51
|
Moodley S, Kroon E, Naidoo CC, Nyawo GR, Wu BG, Naidoo S, Chiyaka TL, Tshivhula H, Singh S, Li Y, Warren RM, Hoal EG, Schurr E, Clemente JC, Segal LN, Möller M, Theron G. Latent Tuberculosis Infection Is Associated with an Enrichment of Short-Chain Fatty Acid-Producing Bacteria in the Stool of Women Living with HIV. Microorganisms 2024; 12:1048. [PMID: 38930430 PMCID: PMC11205370 DOI: 10.3390/microorganisms12061048] [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: 04/12/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 06/28/2024] Open
Abstract
Latent tuberculosis infection (LTBI) is common in people living with HIV (PLHIV) in high-TB-burden settings. Active TB is associated with specific stool taxa; however, little is known about the stool microbiota and LTBI in PLHIV. We characterised the stool microbiota of PLHIV with [interferon-γ release assay (IGRA)- and tuberculin skin test (TST)-positive] or without (IGRA- and TST-negative) LTBI (n = 25 per group). The 16S rRNA DNA sequences were analysed using QIIME2, Dirichlet-Multinomial Mixtures, DESeq2, and PICRUSt2. No α- or β-diversity differences occurred by LTBI status; however, LTBI-positive people were Faecalibacterium-, Blautia-, Gemmiger-, and Bacteroides-enriched and Moryella-, Atopobium-, Corynebacterium-, and Streptococcus-depleted. Inferred metagenome data showed that LTBI-negative-enriched pathways included several metabolite degradation pathways. Stool from LTBI-positive people demonstrated differential taxa abundance based on a quantitative response to antigen stimulation. In LTBI-positive people, older people had different β-diversities than younger people, whereas in LTBI-negative people, no differences occurred across age groups. Amongst female PLHIV, those with LTBI were, vs. those without LTBI, Faecalibacterium-, Blautia-, Gemmiger-, and Bacteriodes-enriched, which are producers of short-chain fatty acids. Taxonomic differences amongst people with LTBI occurred according to quantitative response to antigen stimulation and age. These data enhance our understanding of the microbiome's potential role in LTBI.
Collapse
Affiliation(s)
- Suventha Moodley
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa; (S.M.); (E.K.); (C.C.N.); (G.R.N.); (S.N.); (T.L.C.); (H.T.); (R.M.W.); (E.G.H.); (M.M.)
- African Microbiome Institute, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa
| | - Elouise Kroon
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa; (S.M.); (E.K.); (C.C.N.); (G.R.N.); (S.N.); (T.L.C.); (H.T.); (R.M.W.); (E.G.H.); (M.M.)
| | - Charissa C. Naidoo
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa; (S.M.); (E.K.); (C.C.N.); (G.R.N.); (S.N.); (T.L.C.); (H.T.); (R.M.W.); (E.G.H.); (M.M.)
- African Microbiome Institute, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa
| | - Georgina R. Nyawo
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa; (S.M.); (E.K.); (C.C.N.); (G.R.N.); (S.N.); (T.L.C.); (H.T.); (R.M.W.); (E.G.H.); (M.M.)
- African Microbiome Institute, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa
| | - Benjamin G. Wu
- Division of Pulmonary and Critical Care Medicine, New York University Grossman School of Medicine, NYU Langone Health, New York, NY 10016, USA; (B.G.W.); (S.S.); (Y.L.); (L.N.S.)
| | - Selisha Naidoo
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa; (S.M.); (E.K.); (C.C.N.); (G.R.N.); (S.N.); (T.L.C.); (H.T.); (R.M.W.); (E.G.H.); (M.M.)
| | - Tinaye L. Chiyaka
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa; (S.M.); (E.K.); (C.C.N.); (G.R.N.); (S.N.); (T.L.C.); (H.T.); (R.M.W.); (E.G.H.); (M.M.)
- African Microbiome Institute, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa
| | - Happy Tshivhula
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa; (S.M.); (E.K.); (C.C.N.); (G.R.N.); (S.N.); (T.L.C.); (H.T.); (R.M.W.); (E.G.H.); (M.M.)
- African Microbiome Institute, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa
| | - Shivani Singh
- Division of Pulmonary and Critical Care Medicine, New York University Grossman School of Medicine, NYU Langone Health, New York, NY 10016, USA; (B.G.W.); (S.S.); (Y.L.); (L.N.S.)
| | - Yonghua Li
- Division of Pulmonary and Critical Care Medicine, New York University Grossman School of Medicine, NYU Langone Health, New York, NY 10016, USA; (B.G.W.); (S.S.); (Y.L.); (L.N.S.)
| | - Robin M. Warren
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa; (S.M.); (E.K.); (C.C.N.); (G.R.N.); (S.N.); (T.L.C.); (H.T.); (R.M.W.); (E.G.H.); (M.M.)
| | - Eileen G. Hoal
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa; (S.M.); (E.K.); (C.C.N.); (G.R.N.); (S.N.); (T.L.C.); (H.T.); (R.M.W.); (E.G.H.); (M.M.)
| | - Erwin Schurr
- Department of Biochemistry, McGill University, Montreal, QC H3A 1Y6, Canada;
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, 1001 Boul Décarie, Site Glen Block E, Room EM3.3210, Montréal, QC H4A 3J1, Canada
- McGill International TB Centre, McGill University, Montréal, QC H3A3J1, Canada
- Departments of Medicine and Human Genetics, McGill University, Montréal, QC H3A0C7, Canada
| | - Jose C. Clemente
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Leopoldo N. Segal
- Division of Pulmonary and Critical Care Medicine, New York University Grossman School of Medicine, NYU Langone Health, New York, NY 10016, USA; (B.G.W.); (S.S.); (Y.L.); (L.N.S.)
| | - Marlo Möller
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa; (S.M.); (E.K.); (C.C.N.); (G.R.N.); (S.N.); (T.L.C.); (H.T.); (R.M.W.); (E.G.H.); (M.M.)
| | - Grant Theron
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa; (S.M.); (E.K.); (C.C.N.); (G.R.N.); (S.N.); (T.L.C.); (H.T.); (R.M.W.); (E.G.H.); (M.M.)
- African Microbiome Institute, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa
| |
Collapse
|
52
|
Melilli MG, Buzzanca C, Di Stefano V. Quality characteristics of cereal-based foods enriched with different degree of polymerization inulin: A review. Carbohydr Polym 2024; 332:121918. [PMID: 38431396 DOI: 10.1016/j.carbpol.2024.121918] [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: 11/08/2023] [Revised: 02/02/2024] [Accepted: 02/04/2024] [Indexed: 03/05/2024]
Abstract
Vegetables, cereals and fruit are foods rich in fibre with beneficial and nutritional effects as their consumption reduces the onset of degenerative diseases, especially cardiovascular ones. Among fibres, inulin, oligofructose or fructooligosaccharide (FOS) are the best-studied. Inulin is a generic term to cover all linear β(2-1) fructans, with a variable degree of polymerization. In this review a better understanding of the importance of the degree of polymerization of inulin as a dietary fibre, functions, health benefits, classifications, types and its applications in the food industry was considered in different fortified foods. Inulin has been used to increase the nutritional and healthy properties of the product as a sweetener and as a substitute for fats and carbohydrates, improving the nutritional value and decreasing the glycemic index, with the advantage of not compromising taste and consistency of the product. Bifidogenic and prebiotic effects of inulin have been well established, inulin-type fructans are fermented by the colon to produce short-chain fatty acids, with important local and systemic actions. Addition of inulin with different degrees of polymerization to daily foods for the production of fortified pasta and bread was reviewed, and the impact on sensorial, technological and organoleptic characteristics even of gluten-free bread was also reported.
Collapse
Affiliation(s)
- Maria Grazia Melilli
- National Council of Research, Institute of Biomolecular Chemistry (CNR-ICB), Catania, Italy.
| | - Carla Buzzanca
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Italy.
| | - Vita Di Stefano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Italy; National Biodiversity Future Center (NBFC), 90123, Palermo, Italy.
| |
Collapse
|
53
|
Flint HJ, Louis P, Duncan SH. Why does increased microbial fermentation in the human colon shift toward butyrate? AIMS Microbiol 2024; 10:311-319. [PMID: 38919716 PMCID: PMC11194621 DOI: 10.3934/microbiol.2024016] [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: 03/05/2024] [Revised: 04/15/2024] [Accepted: 04/25/2024] [Indexed: 06/27/2024] Open
Abstract
The microbial community of the human large intestine mainly ferments dietary fiber to short chain fatty acids (SCFAs), which are efficiently absorbed by the host. The three major SCFAs (acetate, propionate, and butyrate) have different fates within the body and different effects on health. A recent analysis of 10 human volunteer studies established that the proportions of these SCFA in fecal samples significantly shifted towards butyrate as the overall concentration of SCFA increased. Butyrate plays a key role in gut health and is preferentially utilized as an energy source by the colonic epithelium. Here we discuss possible mechanisms that underlie this 'butyrate shift'; these include the selection for butyrate-producing bacteria within the microbiota by certain types of fiber, and the possibility of additional butyrate formation from lactate and acetate by metabolite cross-feeding. However, a crucial factor appears to be the pH in the proximal colon, which decreases as the SCFA concentrations increase. A mildly acidic pH has been shown to have an important impact on microbial competition and on the stoichiometry of butyrate production. Understanding these complex interactions has been greatly aided by the refinement of theoretical models of the colonic microbiota that assume a small number (10) of microbial functional groups (MFGs).
Collapse
Affiliation(s)
| | | | - Sylvia H. Duncan
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, UK AB25 2ZD
| |
Collapse
|
54
|
Huang X, Nie S, Fu X, Nan S, Ren X, Li R. Exploring the prebiotic potential of hydrolyzed fucoidan fermented in vitro with human fecal inocula: Impact on microbiota and metabolome. Int J Biol Macromol 2024; 267:131202. [PMID: 38556225 DOI: 10.1016/j.ijbiomac.2024.131202] [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: 01/07/2024] [Revised: 03/17/2024] [Accepted: 03/26/2024] [Indexed: 04/02/2024]
Abstract
Fucoidan is widely applied in food and pharmaceutical industry for the promising bioactivities. Low-molecular weight hydrolyzed fucoidan has gained attention for its beneficial health effects. Here, the modulation on microbiome and metabolome features of fucoidan and its acidolyzed derivatives (HMAF, 1.5-20 kDa; LMAF, <1.5 kDa) were investigated through human fecal cultures. Fucose is the main monosaccharide component in fucoidan and LMAF, while HMAF contains abundant glucuronic acid. LMAF fermentation resulted in the highest production of short-chain fatty acids, with acetate and propionate reaching maximum levels of 13.46 mmol/L and 11.57 mmol/L, respectively. Conversely, HMAF exhibited a maximum butyrate production of 9.28 mmol/L. Both fucoidan and acidolyzed derivatives decreased the abundance of Escherichia-Shigella and Klebsiella in human fecal cultures. Fucoidan and HMAF prefer to improve the abundance of Bacteroides. However, LMAF showed positive influence on Bifidobacterium, Lactobacillus, and Megamonas. Untargeted metabolome indicated that fucoidan and its derivatives mainly altered the metabolic level of lipids, indole, and their derivatives, with fucoidan and HMAF promoting higher level of indole-3-propionic acid and indole-3-carboxaldehyde compared to LMAF. Considering the chemical structural differences, this study suggested that hydrolyzed fucoidan can provide potential therapeutic applications for targeted regulation of microbial communities.
Collapse
Affiliation(s)
- Xinru Huang
- State Key Laboratory of Food Science and Resources, Nanchang University, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang 330047, Jiangxi, People's Republic of China
| | - Shaoping Nie
- State Key Laboratory of Food Science and Resources, Nanchang University, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang 330047, Jiangxi, People's Republic of China
| | - Xiaodan Fu
- State Key Laboratory of Food Science and Resources, Nanchang University, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang 330047, Jiangxi, People's Republic of China.
| | - Shihao Nan
- State Key Laboratory of Food Science and Resources, Nanchang University, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang 330047, Jiangxi, People's Republic of China
| | - Xinmiao Ren
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, People's Republic of China
| | - Rong Li
- Qingdao Women and Children's Hospital, Qingdao 266034, Shandong, People's Republic of China
| |
Collapse
|
55
|
Gleasman-DeSimone S. The Microbiome and Irritable Bowel Syndrome: An Emerging Hope for Treatment. Gastroenterol Nurs 2024; 47:177-184. [PMID: 38847427 DOI: 10.1097/sga.0000000000000813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/08/2024] [Indexed: 06/13/2024] Open
Abstract
Irritable bowel syndrome is a gastrointestinal disorder that affects 15%-20% of the US population. Its symptoms can have negative effects on a person's quality of life, and its treatment can be associated with high medical costs. An emerging area of irritable bowel syndrome research concerns the relationship between this condition and the gut microbiome. The purpose of this article is not only to review irritable bowel syndrome, and the role that the microbiome can play in its symptoms, but also to examine new emerging pathways that could blaze the trail for more individualized treatments. If equipped with this knowledge, gastrointestinal nurses and providers of care can be better prepared to help patients with irritable bowel syndrome in order to manage symptoms and improve their quality of life.
Collapse
Affiliation(s)
- Sara Gleasman-DeSimone
- Sara Gleasman-DeSimone, PhD, RN, NP-C, Le Moyne College Nursing Department, Syracuse, New York
| |
Collapse
|
56
|
Liu D, Xie LS, Lian S, Li K, Yang Y, Wang WZ, Hu S, Liu SJ, Liu C, He Z. Anaerostipes hadrus, a butyrate-producing bacterium capable of metabolizing 5-fluorouracil. mSphere 2024; 9:e0081623. [PMID: 38470044 PMCID: PMC11036815 DOI: 10.1128/msphere.00816-23] [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: 12/27/2023] [Accepted: 02/22/2024] [Indexed: 03/13/2024] Open
Abstract
Anaerostipes hadrus (A. hadrus) is a dominant species in the human gut microbiota and considered a beneficial bacterium for producing probiotic butyrate. However, recent studies have suggested that A. hadrus may negatively affect the host through synthesizing fatty acid and metabolizing the anticancer drug 5-fluorouracil, indicating that the impact of A. hadrus is complex and unclear. Therefore, comprehensive genomic studies on A. hadrus need to be performed. We integrated 527 high-quality public A. hadrus genomes and five distinct metagenomic cohorts. We analyzed these data using the approaches of comparative genomics, metagenomics, and protein structure prediction. We also performed validations with culture-based in vitro assays. We constructed the first large-scale pan-genome of A. hadrus (n = 527) and identified 5-fluorouracil metabolism genes as ubiquitous in A. hadrus genomes as butyrate-producing genes. Metagenomic analysis revealed the wide and stable distribution of A. hadrus in healthy individuals, patients with inflammatory bowel disease, and patients with colorectal cancer, with healthy individuals carrying more A. hadrus. The predicted high-quality protein structure indicated that A. hadrus might metabolize 5-fluorouracil by producing bacterial dihydropyrimidine dehydrogenase (encoded by the preTA operon). Through in vitro assays, we validated the short-chain fatty acid production and 5-fluorouracil metabolism abilities of A. hadrus. We observed for the first time that A. hadrus can convert 5-fluorouracil to α-fluoro-β-ureidopropionic acid, which may result from the combined action of the preTA operon and adjacent hydA (encoding bacterial dihydropyrimidinase). Our results offer novel understandings of A. hadrus, exceptionally functional features, and potential applications. IMPORTANCE This work provides new insights into the evolutionary relationships, functional characteristics, prevalence, and potential applications of Anaerostipes hadrus.
Collapse
Affiliation(s)
- Danping Liu
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology of the People’s Republic of China, Beijing, China
- Key Laboratory of Biomechanics and Mechanobiology, Beihang University, Ministry of Education, Beijing, China
| | - Li-Sheng Xie
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Shitao Lian
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology of the People’s Republic of China, Beijing, China
- Key Laboratory of Biomechanics and Mechanobiology, Beihang University, Ministry of Education, Beijing, China
| | - Kexin Li
- Systems Biology and Bioinformatics (SBI), Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Yun Yang
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology of the People’s Republic of China, Beijing, China
- Key Laboratory of Biomechanics and Mechanobiology, Beihang University, Ministry of Education, Beijing, China
| | - Wen-Zhao Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Songnian Hu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Shuang-Jiang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Chang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Zilong He
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology of the People’s Republic of China, Beijing, China
- Key Laboratory of Biomechanics and Mechanobiology, Beihang University, Ministry of Education, Beijing, China
| |
Collapse
|
57
|
Liu H, Wang S, Chen M, Ji H, Zhang D. Effects of Lactobacillus-fermented low-protein diets on the growth performance, nitrogen excretion, fecal microbiota and metabolomic profiles of finishing pigs. Sci Rep 2024; 14:8612. [PMID: 38616198 PMCID: PMC11016537 DOI: 10.1038/s41598-024-58832-y] [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: 01/05/2024] [Accepted: 04/03/2024] [Indexed: 04/16/2024] Open
Abstract
This study investigated the effects of Lactobacillus-fermented low-protein diet on the growth performance, nitrogen balance, fecal microbiota, and metabolomic profiles of finishing pigs. A total of 90 finishing pigs were assigned to one of three dietary treatments including a normal protein diet (CON) as well as two experimental diets in which a low-protein diet supplemented with 0 (LP) or 1% Lactobacillus-fermented low-protein feed (FLP). In comparison with CON, the LP and FLP significantly increased average daily gain (P = 0.044), significantly decreased feed to gain ratio (P = 0.021), fecal nitrogen (P < 0.01), urine nitrogen (P < 0.01), and total nitrogen (P < 0.01), respectively. The LP group exhibited increased abundances of unclassified_f_Selenomonadaceae, Coprococcus, Faecalibacterium, and Butyricicoccus, while the abundances of Verrucomicrobiae, Verrucomicrobiales, Akkermansiaceae, and Akkermansia were enriched in the FLP group. Low-protein diet-induced metabolic changes were enriched in sesquiterpenoid and triterpenoid biosynthesis and Lactobacillus-fermented low-protein feed-induced metabolic changes were enriched in phenylpropanoid biosynthesis and arginine biosynthesis. Overall, low-protein diet and Lactobacillus-fermented low-protein diet improved the growth performance and reduce nitrogen excretion, possibly via altering the fecal microbiota and metabolites in the finishing pigs. The present study provides novel ideas regarding the application of the low-protein diet and Lactobacillus-fermented low-protein diet in swine production.
Collapse
Affiliation(s)
- Hui Liu
- Institute of Animal Science and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China
| | - Sixin Wang
- Institute of Animal Science and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China
| | - Meixia Chen
- Institute of Animal Science and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China
| | - Haifeng Ji
- Institute of Animal Science and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China.
| | - Dongyan Zhang
- Institute of Animal Science and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China.
| |
Collapse
|
58
|
Wang X, Li Y, Wang X, Wang R, Hao Y, Ren F, Wang P, Fang B. Faecalibacterium prausnitzii Supplementation Prevents Intestinal Barrier Injury and Gut Microflora Dysbiosis Induced by Sleep Deprivation. Nutrients 2024; 16:1100. [PMID: 38674791 PMCID: PMC11054126 DOI: 10.3390/nu16081100] [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: 02/24/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Sleep deprivation (SD) leads to impaired intestinal barrier function and intestinal flora disorder, especially a reduction in the abundance of the next generation of probiotic Faecalibacterium prausnitzii (F. prausnitzii). However, it remains largely unclear whether F. prausnitzii can ameliorate SD-induced intestinal barrier damage. A 72 h SD mouse model was used in this research, with or without the addition of F. prausnitzii. The findings indicated that pre-colonization with F. prausnitzii could protect against tissue damage from SD, enhance goblet cell count and MUC2 levels in the colon, boost tight-junction protein expression, decrease macrophage infiltration, suppress pro-inflammatory cytokine expression, and reduce apoptosis. We found that the presence of F. prausnitzii helped to balance the gut microbiota in SD mice by reducing harmful bacteria like Klebsiella and Staphylococcus, while increasing beneficial bacteria such as Akkermansia. Ion chromatography analysis revealed that F. prausnitzii pretreatment increased the fecal butyrate level in SD mice. Overall, these results suggested that incorporating F. prausnitzii could help reduce gut damage caused by SD, potentially by enhancing the intestinal barrier and balancing gut microflora. This provides a foundation for utilizing probiotics to protect against intestinal illnesses.
Collapse
Affiliation(s)
- Xintong Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (X.W.); (Y.L.); (R.W.); (Y.H.); (F.R.)
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (X.W.); (Y.L.); (R.W.); (Y.H.); (F.R.)
| | - Xifan Wang
- Department of Obstetrics and Gynecology, Columbia University, New York, NY 10032, USA;
| | - Ran Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (X.W.); (Y.L.); (R.W.); (Y.H.); (F.R.)
| | - Yanling Hao
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (X.W.); (Y.L.); (R.W.); (Y.H.); (F.R.)
| | - Fazheng Ren
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (X.W.); (Y.L.); (R.W.); (Y.H.); (F.R.)
- Food Laboratory of Zhongyuan, Luohe 462000, China
| | - Pengjie Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (X.W.); (Y.L.); (R.W.); (Y.H.); (F.R.)
- Food Laboratory of Zhongyuan, Luohe 462000, China
| | - Bing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (X.W.); (Y.L.); (R.W.); (Y.H.); (F.R.)
| |
Collapse
|
59
|
Imdad S, Kim JH, So B, Jang J, Park J, Lim W, Lee YK, Shin WS, Hillyer T, Kang C. Effect of aerobic exercise and particulate matter exposure duration on the diversity of gut microbiota. Anim Cells Syst (Seoul) 2024; 28:137-151. [PMID: 38601060 PMCID: PMC11005883 DOI: 10.1080/19768354.2024.2338855] [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: 11/14/2023] [Accepted: 03/31/2024] [Indexed: 04/12/2024] Open
Abstract
Inhalation of ambient particulate matter (PM) can disrupt the gut microbiome, while exercise independently influences the gut microbiome by promoting beneficial bacteria. In this study, we analyzed changes in gut microbial diversity and composition in response to combined interventions of PM exposure and aerobic exercise, extending up to 12 weeks. This investigation was conducted using mice, categorized into five groups: control group (Con), exercise group (EXE), exercise group followed by 3-day exposure to PM (EXE + 3-day PM), particulate matter exposure (PM), and PM exposure with concurrent treadmill exercise (PME). Notably, the PM group exhibited markedly lower alpha diversity and richness compared to the Con group and our analysis of beta diversity revealed significant variations among the intervention groups. Members of the Lachnospiraceae family showed significant enhancement in the exercise intervention groups (EXE and PME) compared to the Con and PM groups. The biomarker Lactobacillus, Coriobacteraceae, and Anaerofustis were enriched in the EXE group, while Desulfovibrionaceae, Mucispirillum schaedleri, Lactococcus and Anaeroplasma were highly enriched in the PM group. Differential abundance analysis revealed that Paraprevotella, Bacteroides, and Blautia were less abundant in the 12-week PM exposure group than in the 3-day PM exposure group. Moreover, both the 3-day and 12-week PM exposure groups exhibited a reduced relative abundance of Bacteroides uniformis, SMB53, and Staphylococcus compared to non-PM exposure groups. These findings will help delineate the possible roles and associations of altered microbiota resulting from the studied interventions, paving the way for future mechanistic research.
Collapse
Affiliation(s)
- Saba Imdad
- Laboratory of Molecular Metabolism in Health & Disease, Sport Science Research Institute, Inha University, Incheon, South Korea
- Department of Biomedical Laboratory Science, College of Health Science, Cheongju University, Cheongju, South Korea
| | - Jin-Hee Kim
- Department of Biomedical Laboratory Science, College of Health Science, Cheongju University, Cheongju, South Korea
| | - Byunghun So
- Laboratory of Molecular Metabolism in Health & Disease, Sport Science Research Institute, Inha University, Incheon, South Korea
| | - Junho Jang
- Laboratory of Molecular Metabolism in Health & Disease, Sport Science Research Institute, Inha University, Incheon, South Korea
| | - Jinhan Park
- Laboratory of Molecular Metabolism in Health & Disease, Sport Science Research Institute, Inha University, Incheon, South Korea
| | - Wonchung Lim
- Department of Sports Medicine, College of Health Science, Cheongju University, Cheongju, South Korea
| | - Yoon-Kwang Lee
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Woo Shik Shin
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Trae Hillyer
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Chounghun Kang
- Laboratory of Molecular Metabolism in Health & Disease, Sport Science Research Institute, Inha University, Incheon, South Korea
- Department of Physical Education, College of Education, Inha University, Incheon, South Korea
| |
Collapse
|
60
|
Zhao M, Zhang Y, Li Y, Liu K, Bao K, Li G. Impact of Pediococcus acidilactici GLP06 supplementation on gut microbes and metabolites in adult beagles: a comparative analysis. Front Microbiol 2024; 15:1369402. [PMID: 38633690 PMCID: PMC11021720 DOI: 10.3389/fmicb.2024.1369402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/13/2024] [Indexed: 04/19/2024] Open
Abstract
There is growing interest in the potential health benefits of probiotics for both humans and animals. The study aimed to investigate the effects of feeding the canine-derived probiotic Pediococcus acidilactici GLP06 to adult beagles by analysing the microbiome and metabolome. Twenty-four healthy adult beagles were randomly assigned to four groups. The CK group received a standard diet, while the three probiotic groups, the LG group (2 × 108 CFU/day/dog), MG group (2 × 109 CFU/day/dog), and HG group (2 × 1010 CFU/day/dog), received the standard diet supplemented with varying amounts of probiotics. The results show that, compared to the CK group, total antioxidant capacity was significantly increased in the MG and HG groups (p < 0.05), and superoxide dismutase and catalase were significantly increased in the HG group (p < 0.05). Compared to the CK group, malondialdehyde and blood urea nitrogen content were significantly decreased in the MG and HG groups (p < 0.05). Additionally, secretory immunoglobulin A activity was significantly increased in the HG group compared to the CK and LG groups (p < 0.05), and immunoglobulin G activity was significantly increased in the HG group compared to the CK, LG, and MG groups (p < 0.05). In addition, compared with the CK group, the abundance of Faecalitalea and Collinsella increased in the LG group, and the relative abundance of Tyzzerella and Parasutterella increased in the MG group. The α diversity and the relative abundances of beneficial bacteria (Faecalibacterium, Lachnospiraceae_NK4A1316, and Ruminococcaceae_UCG-005) were higher in the HG group than in the CK group. Furthermore, acetic acid content was significantly increased in the HG group compared to the CK, LG, and MG groups (p < 0.05). Butyric acid, isobutyric acid, and the total SCFA content were significantly increased in the HG group compared to the CK group (p < 0.05). Moreover, metabolome analysis revealed 111 upregulated and 171 downregulated metabolites in the HG group. In conclusion, this study presents evidence that supplementing with P. acidilactici GLP06 can have a positive impact on antioxidant activity, immunoproteins, SCFAs, and gut microbiota in adult beagles. These findings highlight the potential of probiotics as a dietary intervention to enhance gut health and overall wellbeing in companion animals.
Collapse
Affiliation(s)
- Mengdi Zhao
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
- College of Animal Science and Technology, Jilin Agriculture University, Changchun, China
| | - Yuanyuan Zhang
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| | - Yueyao Li
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| | - Keyuan Liu
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| | - Kun Bao
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| | - Guangyu Li
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
61
|
Moodley S, Kroon E, Naidoo CC, Nyawo GR, Wu BG, Naidoo S, Chiyaka TL, Tshivhula H, Singh S, Li Y, Warren RM, Hoal EG, Schurr E, Clemente J, Segal LN, Möller M, Theron G. Latent tuberculosis infection is associated with an enrichment of short chain fatty acid producing bacteria in the stool of women living with HIV. RESEARCH SQUARE 2024:rs.3.rs-4182285. [PMID: 38645218 PMCID: PMC11030539 DOI: 10.21203/rs.3.rs-4182285/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Background Latent tuberculosis infection (LTBI) is common in people living with HIV (PLHIV) in high TB burden settings. Active TB is associated with specific stool taxa; however, little is known about the stool microbiota and LTBI, including in PLHIV. Method Within a parent study that recruited adult females with HIV from Cape Town, South Africa into predefined age categories (18-25, 35-60 years), we characterised the stool microbiota of those with [interferon-γ release assay (IGRA)- and tuberculin skin test (TST)-positive] or without (IGRA- and TST- negative) LTBI (n=25 per group). 16S rRNA DNA sequences were analysed using QIIME2, Dirichlet Multinomial Mixtures, DESeq2 and PICRUSt2. Results No α- or β-diversity differences occurred by LTBI status; however, LTBI-positives were Faecalibacterium-, Blautia-, Gemmiger-, Bacteroides-enriched and Moryella-, Atopobium-, Corynebacterium-, Streptococcus-depleted. Inferred metagenome data showed LTBI-negative-enriched pathways included several involved in methylglyoxal degradation, L-arginine, putrescine, 4-aminobutanoate degradation and L-arginine and ornithine degradation. Stool from LTBI-positives demonstrated differential taxa abundance based on a quantitative response to antigen stimulation (Acidaminococcus-enrichment and Megamonas-, Alistipes-, and Paraprevotella-depletion associated with higher IGRA or TST responses, respectively). In LTBI-positives, older people had different β-diversities than younger people whereas, in LTBI-negatives, no differences occurred across age groups. Conclusion Amongst female PLHIV, those with LTBI had, vs. those without LTBI, Faecalibacterium, Blautia, Gemmiger, Bacteriodes-enriched, which are producers of short chain fatty acids. Taxonomic differences amongst people with LTBI occurred according to quantitative response to antigen stimulation and age. These data enhance our understanding of the microbiome's potential role in LTBI.
Collapse
|
62
|
Chen J, Yin J, Xie H, Lu W, Wang H, Zhao J, Zhu J. Mannan-oligosaccharides promote gut microecological recovery after antibiotic disturbance. Food Funct 2024; 15:3810-3823. [PMID: 38511344 DOI: 10.1039/d4fo00332b] [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: 03/22/2024]
Abstract
Antibiotic treatment often causes collateral damage to the gut microbiota, including changes in its diversity and composition. Dietary fiber helps maintain intestinal health, regulate short-chain fatty acids, and promote the recovery of the intestinal microbiome. However, it is currently unknown which specific plant-based dietary fiber is optimal as a dietary supplement for restoring the intestinal microbiota after antibiotic disturbance. Previously, we proposed predictive recovery-associated bacterial species (p-RABs) and identified the most important interventions. This study aimed to identify an optimal form of dietary fiber to recover the gut microbiome after antibiotic treatment. Therefore, we examined the types of dietary fibers associated with p-RABs through a p-RAB-metabolite bilayer network constructed from prior knowledge; we searched for dietary fiber that could provide nutritional support for Akkermansia muciniphila and Bacteroides uniformis. C57BL/6J mice were fed with 500 mg kg-1 of different types of dietary fibers daily for one week after being treated with ampicillin. The results showed that mannan-oligosaccharides could better promote the diversity of intestinal microbial growth, enhance the recovery of most genera, including Akkermansia and Bacteroides, and inhibit certain pathogenic bacteria, such as Proteus, compared to the other fiber types. Furthermore, mannan-oligosaccharides could regulate the levels of short-chain fatty acids, especially butyric acid. Functional predictions showed that starch metabolism, galactose metabolism, and the metabolism of other carbohydrates played key roles in the early recovery process. In conclusion, mannan-oligosaccharides could enhance the recovery of the intestinal microbiome after antibiotic treatment, offering valuable insights for targeted dietary strategies.
Collapse
Affiliation(s)
- Jing Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Jialin Yin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Heqiang Xie
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
- International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, China
| | - Hongchao Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, China
| | - Jinlin Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
63
|
Song I, Yang J, Saito M, Hartanto T, Nakayama Y, Ichinohe T, Fukuda S. Prebiotic inulin ameliorates SARS-CoV-2 infection in hamsters by modulating the gut microbiome. NPJ Sci Food 2024; 8:18. [PMID: 38485724 PMCID: PMC10940623 DOI: 10.1038/s41538-024-00248-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 01/16/2024] [Indexed: 03/18/2024] Open
Abstract
Current treatment options for COVID-19 are limited, with many antivirals and immunomodulators restricted to the most severe cases and preventative care limited to vaccination. As the SARS-CoV-2 virus and its increasing variants threaten to become a permanent fixture of our lives, this new reality necessitates the development of cost-effective and accessible treatment options for COVID-19. Studies have shown that there are correlations between the gut microbiome and severity of COVID-19, especially with regards to production of physiologically beneficial short-chain fatty acids (SCFAs) by gut microbes. In this study, we used a Syrian hamster model to study how dietary consumption of the prebiotic inulin affected morbidity and mortality resulting from SARS-CoV-2 infection. After two weeks of observation, we discovered that inulin supplementation attenuated morbid weight loss and increased survival rate in hamster subjects. An analysis of microbiome community structure showed significant alterations in 15 genera. Notably, there were also small increases in fecal DCA and a significant increase in serum DCA, perhaps highlighting a role for this secondary bile acid in conferring protection against SARS-CoV-2. In light of these results, inulin and other prebiotics are promising targets for future investigation as preventative treatment options for COVID-19.
Collapse
Affiliation(s)
- Isaiah Song
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Jiayue Yang
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Misa Saito
- Metagen, Inc., Tsuruoka, Yamagata, Japan
| | | | | | - Takeshi Ichinohe
- Division of Viral Infection, Department of Infectious Disease Control, International Research Center for Infectious Diseases, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan.
- Metagen, Inc., Tsuruoka, Yamagata, Japan.
- Gut Environmental Design Group, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Kanagawa, Japan.
- Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan.
- Laboratory for Regenerative Microbiology, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
64
|
Elango A, Nesam VD, Sukumar P, Lawrence I, Radhakrishnan A. Postbiotic butyrate: role and its effects for being a potential drug and biomarker to pancreatic cancer. Arch Microbiol 2024; 206:156. [PMID: 38480544 DOI: 10.1007/s00203-024-03914-8] [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: 01/19/2024] [Revised: 02/17/2024] [Accepted: 02/26/2024] [Indexed: 04/16/2024]
Abstract
Postbiotics are produced by microbes and have recently gained importance in the field of oncology due to their beneficial effects to the host, effectiveness against cancer cells, and their ability to suppress inflammation. In particular, butyrate dominates over all other postbiotics both in quantity and anticancer properties. Pancreatic cancer (PC), being one of the most malignant and lethal cancers, reported a decreased 5-year survival rate in less than 10% of the patients. PC causes an increased mortality rate due to its inability to be detected at an early stage but still a promising strategy for its diagnosis has not been achieved yet. It is necessary to diagnose Pancreatic cancer before the metastatic progression stage. The available blood biomarkers lack accurate and proficient diagnostic results. Postbiotic butyrate is produced by gut microbiota such as Rhuminococcus and Faecalibacterium it is involved in cell signalling pathways, autophagy, and cell cycle regulation, and reduction in butyrate concentration is associated with the occurrence of pancreatic cancer. The postbiotic butyrate is a potential biomarker that could detect PC at an early stage, before the metastatic progression stage. Thus, this review focused on the gut microbiota butyrate's role in pancreatic cancer and the immuno-suppressive environment, its effects on histone deacetylase and other immune cells, microbes in major butyrate synthesis pathways, current biomarkers in use for Pancreatic Cancer.
Collapse
Affiliation(s)
- Abinaya Elango
- Department of Pharmacology, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Chengalpattu, Tamil Nadu, 603103, India
| | - Vineeta Debbie Nesam
- Department of Pharmacology, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Chengalpattu, Tamil Nadu, 603103, India
| | - Padmaja Sukumar
- Department of Pharmacology, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Chengalpattu, Tamil Nadu, 603103, India
| | - Infancia Lawrence
- Priyadharshani Research and Development, Kelambakkam, Chengalpattu, Tamil Nadu, 603103, India
| | - Arunkumar Radhakrishnan
- Department of Pharmacology, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Chengalpattu, Tamil Nadu, 603103, India.
| |
Collapse
|
65
|
Shearer J, Shah S, MacInnis MJ, Shen-Tu G, Mu C. Dose-Responsive Effects of Iron Supplementation on the Gut Microbiota in Middle-Aged Women. Nutrients 2024; 16:786. [PMID: 38542697 PMCID: PMC10975138 DOI: 10.3390/nu16060786] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 11/12/2024] Open
Abstract
Oral iron supplementation is the first-line treatment for addressing iron deficiency, a concern particularly relevant to women who are susceptible to sub-optimal iron levels. Nevertheless, the impact of iron supplementation on the gut microbiota of middle-aged women remains unclear. To investigate the association between iron supplementation and the gut microbiota, healthy females aged 40-65 years (n = 56, BMI = 23 ± 2.6 kg/m2) were retrospectively analyzed from the Alberta's Tomorrow Project. Fecal samples along with various lifestyle, diet, and health questionnaires were obtained. The gut microbiota was assessed by 16S rRNA sequencing. Individuals were matched by age and BMI and classified as either taking no iron supplement, a low-dose iron supplement (6-10 mg iron/day), or high-dose iron (>100 mg/day). Compositional and functional analyses of microbiome data in relation to iron supplementation were investigated using various bioinformatics tools. Results revealed that iron supplementation had a dose-dependent effect on microbial communities. Elevated iron intake (>100 mg) was associated with an augmentation of Proteobacteria and a reduction in various taxa, including Akkermansia, Butyricicoccus, Verrucomicrobia, Ruminococcus, Alistipes, and Faecalibacterium. Metagenomic prediction further suggested the upregulation of iron acquisition and siderophore biosynthesis following high iron intake. In conclusion, adequate iron levels are essential for the overall health and wellbeing of women through their various life stages. Our findings offer insights into the complex relationships between iron supplementation and the gut microbiota in middle-aged women and underscore the significance of iron dosage in maintaining optimal gut health.
Collapse
Affiliation(s)
- Jane Shearer
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada; (J.S.); (S.S.); (M.J.M.)
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Shrushti Shah
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada; (J.S.); (S.S.); (M.J.M.)
| | - Martin J. MacInnis
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada; (J.S.); (S.S.); (M.J.M.)
| | - Grace Shen-Tu
- Alberta’s Tomorrow Project, Cancer Control Alberta, Alberta Health Services, Calgary, AB T2T 5C7, Canada;
| | - Chunlong Mu
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
66
|
Bermudez C, Yao H, Widaningrum, Williams BA, Flanagan BM, Gidley MJ, Mikkelsen D. Biomass attachment and microbiota shifts during porcine faecal in vitro fermentation of almond and macadamia nuts differing in particle sizes. Food Funct 2024; 15:2406-2421. [PMID: 38265095 DOI: 10.1039/d3fo03612j] [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: 01/25/2024]
Abstract
Nuts are highly nutritious and good sources of dietary fibre, when consumed as part of a healthy human diet. Upon consumption, nut particles of various sizes containing lipids entrapped by the plant cell walls enter the large intestine where they are fermented by the resident microbiota. This study investigated the microbial community shifts during in vitro fermentation of almond and macadamia substrates, of two particle sizes including fine particles (F = 250-500 μm) and cell clusters (CC = 710-1000 μm). The aim was to determine how particle size and biomass attachment altered the microbiota. Over the 48 h fermentation duration, short chain fatty acid concentrations increased due to particle size rather than nut type (almond or macadamia). However, nut type did change microbial population dynamics by stimulating specific genera. Tyzzerella, p253418B5 gut group, Lachnospiraceae UCG001, Geotrichum, Enterococcus, Amnipila and Acetitomaculum genera were unique for almonds. For macadamia, three unique genera including Prevotellaceae UCG004, Candidatus Methanomethylophilus and Alistipes were noted. Distinct shifts in the attached microbial biomass were noted due to nut particle size. Bacterial attachment to nut particles was visualised in situ during fermentation, revealing a decrease in lipids and an increase in attached bacteria over time. This interaction may be a pre-requisite for lipid breakdown during nut particle disappearance. Overall, this study provides insights into how nut fermentation alters the gut microbiota and the possible role that gut microbes have in lipid degradation.
Collapse
Affiliation(s)
- Cindy Bermudez
- School of Agriculture and Food Sustainability, The University of Queensland, Brisbane, Australia.
- Centre for Nutrition and Food Sciences (CNAFS), Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Brisbane, Australia
| | - Hong Yao
- Centre for Nutrition and Food Sciences (CNAFS), Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Brisbane, Australia
| | - Widaningrum
- School of Agriculture and Food Sustainability, The University of Queensland, Brisbane, Australia.
- Research Centre for Agroindustry, National Research and Innovation Agency (BRIN), Soekarno Integrated Science Center, Bogor, Indonesia
| | - Barbara A Williams
- Centre for Nutrition and Food Sciences (CNAFS), Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Brisbane, Australia
| | - Bernadine M Flanagan
- Centre for Nutrition and Food Sciences (CNAFS), Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Brisbane, Australia
| | - Michael J Gidley
- Centre for Nutrition and Food Sciences (CNAFS), Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Brisbane, Australia
| | - Deirdre Mikkelsen
- School of Agriculture and Food Sustainability, The University of Queensland, Brisbane, Australia.
- Centre for Nutrition and Food Sciences (CNAFS), Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Brisbane, Australia
| |
Collapse
|
67
|
Bai Y, Zhang Y, Chao C, Yu J, Zhao J, Han D, Wang J, Wang S. Molecular Mechanisms Underlying the Effects of Small Intestinal Fermentation on Enhancement of Prebiotic Characteristics of Cellulose in the Large Intestine. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:3596-3605. [PMID: 38270580 DOI: 10.1021/acs.jafc.3c09146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Knowledge about the prebiotic characteristics of cellulose by in vitro fermentation is not complete due to the neglect of small intestinal fermentation. This study investigated the effects of small intestinal fermentation on the prebiotic characteristics of cellulose in the large intestine and potential mechanisms through an approach of combined in vivo small intestinal fermentation and in vitro fermentation. The structural similarity between cellulose in feces and after processing by the approach of this study confirmed the validity of the approach employed. Results showed that small intestinal fermentation of cellulose increased both acetate and propionate content and enriched Corynebacterium selectively. Compared to in vitro fermentation after in vitro digestion of cellulose, the in vitro fermentation of cellulose after in vivo small intestinal fermentation produced higher contents of acetate and propionate as well as the abundance of probiotics like Ruminococcaceae_UCG-002, Blautia, and Bifidobaterium. The changes in the structural features of cellulose after in vivo small intestinal fermentation were more obvious than those after in vitro digestion, which may account for the greater production of short-chain fatty acids (SCFAs) and the abundance of probiotics. In summary, small intestinal fermentation enhanced the prebiotic characteristics of cellulose in the large intestine by predisrupting its structure.
Collapse
Affiliation(s)
- Yu Bai
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Yiming Zhang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
- College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Chen Chao
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
- College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Jinglin Yu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Jinbiao Zhao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shujun Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
- College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| |
Collapse
|
68
|
Zhang Z, Huang J, Li C, Zhao Z, Cui Y, Yuan X, Wang X, Liu Y, Zhou Y, Zhu Z. The gut microbiota contributes to the infection of bovine viral diarrhea virus in mice. J Virol 2024; 98:e0203523. [PMID: 38299844 PMCID: PMC10878277 DOI: 10.1128/jvi.02035-23] [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: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 02/02/2024] Open
Abstract
Bovine viral diarrhea virus (BVDV) is prevalent worldwide and causes significant economic losses. Gut microbiota is a large microbial community and has a variety of biological functions. However, whether there is a correlation between gut microbiota and BVDV infection and what kind of relation between them have not been reported. Here, we found that gut microbiota composition changed in normal mice after infecting with BVDV, but mainly the low abundance microbe was affected. Interestingly, BVDV infection significantly reduced the diversity of gut microbiota and changed its composition in gut microbiota-dysbiosis mice. Furthermore, compared with normal mice of BVDV infection, there were more viral loads in the duodenum, jejunum, spleen, and liver of the gut microbiota-dysbiosis mice. However, feces microbiota transplantation (FMT) reversed these effects. The data above indicated that the dysbiosis of gut microbiota was a key factor in the high infection rate of BVDV. It is found that the IFN-I signal was involved by investigating the underlying mechanisms. The inhibition of the proliferation and increase in the apoptosis of peripheral blood lymphocytes (PBL) were also observed. However, FMT treatment reversed these changes by regulating PI3K/Akt, ERK, and Caspase-9/Caspase-3 pathways. Furthermore, the involvement of butyrate in the pathogenesis of BVDV was also further confirmed. Our results showed for the first time that gut microbiota acts as a key endogenous defense mechanism against BVDV infection; moreover, targeting regulation of gut microbiota structure and abundance may serve as a new strategy to prevent and control the disease.IMPORTANCEWhether the high infection rate of BVDV is related to gut microbiota has not been reported. In addition, most studies on BVDV focus on in vitro experiments, which limits the study of its prevention and control strategy and its pathogenic mechanism. In this study, we successfully confirmed the causal relationship between gut microbiota and BVDV infection as well as the potential molecular mechanism based on a mouse model of BVDV infection and a mouse model of gut microbiota dysbiosis. Meanwhile, a mouse model which is more susceptible to BVDV provided in this study lays an important foundation for further research on prevention and control strategy of BVDV and its pathogenesis. In addition, the antiviral effect of butyrate, the metabolites of butyrate-producing bacteria, has been further revealed. Overall, our findings provide a promising prevention and control strategy to treat this infectious disease which is distributed worldwide.
Collapse
Affiliation(s)
- Zecai Zhang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
- Engineering Research Center for Prevention and Control of Cattle Diseases, Heilongjiang Province, Daqing, China
- Heilongjiang Province Cultivating Collaborative Innovation Center for The Beidahuang Modern Agricultural Industry Technology, Daqing, China
| | - Jiang Huang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Agriculture and Rural Bureau of Sinan County, Sinan County, Guizhou, China
- Animal Health Supervision Institute of Sinan County, Sinan County, Guizhou, China
| | - Chuang Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
| | - Zhicheng Zhao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
| | - Yueqi Cui
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
| | - Xueying Yuan
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
| | - Xue Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
| | - Yu Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
- Engineering Research Center for Prevention and Control of Cattle Diseases, Heilongjiang Province, Daqing, China
- Heilongjiang Province Cultivating Collaborative Innovation Center for The Beidahuang Modern Agricultural Industry Technology, Daqing, China
| | - Yulong Zhou
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
- Engineering Research Center for Prevention and Control of Cattle Diseases, Heilongjiang Province, Daqing, China
- Heilongjiang Province Cultivating Collaborative Innovation Center for The Beidahuang Modern Agricultural Industry Technology, Daqing, China
| | - Zhanbo Zhu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
- Engineering Research Center for Prevention and Control of Cattle Diseases, Heilongjiang Province, Daqing, China
- Heilongjiang Province Cultivating Collaborative Innovation Center for The Beidahuang Modern Agricultural Industry Technology, Daqing, China
| |
Collapse
|
69
|
Kumar M, Muthurayar T, Karthika S, Gayathri S, Varalakshmi P, Ashokkumar B. Anti-Diabetic Potentials of Lactobacillus Strains by Modulating Gut Microbiota Structure and β-Cells Regeneration in the Pancreatic Islets of Alloxan-Induced Diabetic Rats. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10221-7. [PMID: 38329697 DOI: 10.1007/s12602-024-10221-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2024] [Indexed: 02/09/2024]
Abstract
Diabetes mellitus, a most common endocrine disorder of glucose metabolism, has become a global epidemic and poses a serious public health threat with an increased socio-economic burden. Escalating incidence of diabetes is correlated with changes in lifestyle and food habits that cause gut microbiome dysbiosis and β-cells damage, which can be addressed with dietary interventions containing probiotics. Hence, the search for probiotics of human origin with anti-diabetic, anti-AGE, and anti-ACE potentials has gained renewed interest for the effective management of diabetes and its associated complications. The present study used an alloxan (AXN)-induced diabetic rat model to investigate the effects of potential probiotic Lacticaseibacillus casei MKU1, Lactiplantibacillus pentosus MKU3, and Lactiplantibacillus plantarum MKU7 administration individually on physiochemical parameters related to diabetic pathogenesis. Experimental animals were randomly allotted into six groups viz. NCG (control), DCG (AXN), DGM (metformin), DGP1 (MKU1), DGP2 (MKU3), and DGP3 (MKU7), and biochemical data like serum glucose, insulin, AngII, ACE, HbA1c, and TNF-α levels were measured until 90 days. Our results suggest that oral administration with MKU1, MKU3, or MKU7 significantly improved serum insulin levels, glycemic control, glucose tolerance, and body weight. Additionally, β-cell mass was increased by preserving islet integrity in Lactobacillus-treated diabetic rats, whereas TNF-α (~40%), AngII (~30%), and ACE levels (~50%) were strongly inhibited and enhanced sIgA production (5.8 folds) abundantly. Furthermore, Lactobacillus administration positively influenced the gut microbiome with a significant increase in the abundance of Lactobacillus species and the beneficial Bacteroides uniformis and Bacteroides fragilis, while decreased the pathogenic Proteus vulgaris and Parabacteroides distasonis. Among the probiotic treatment groups, L. pentosus MKU3 performed greatly in almost all parameters, indicating its potential use for alleviating diabetes-associated complications.
Collapse
Affiliation(s)
- Manoj Kumar
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Tharmar Muthurayar
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Sukumaran Karthika
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Santhalingam Gayathri
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Perumal Varalakshmi
- Department of Molecular Microbiology, School of Biotechnology, Madurai Kamaraj University, Madurai, India
| | - Balasubramaniem Ashokkumar
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India.
| |
Collapse
|
70
|
Almeida PP, Da Cruz BO, Thomasi B, Menezes ÁC, Brito ML, Costa NDS, Ito RVA, Degani VAN, Daleprane JB, Magliano DC, Tavares-Gomes AL, Stockler-Pinto MB. Brazil Nut-Enriched Diet Modulates Enteric Glial Cells and Gut Microbiota in an Experimental Model of Chronic Kidney Disease. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2024; 43:201-212. [PMID: 37611162 DOI: 10.1080/27697061.2023.2247057] [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: 01/13/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/25/2023]
Abstract
Introduction: Chronic kidney disease (CKD) promotes gut dysbiosis, and enteric glial reactivity, a feature of intestinal inflammation. Brazil nut modulated enteric glial profile in healthy animals and could modulate these cells in 5/6 nephrectomized rats.Methods: A 5/6 nephrectomy-induced CKD and Sham-operated rats were divided as follows: CKD and Sham received a standard diet and CKD-BN and Sham-BN received a 5% Brazil nut enriched-diet. The protein content of glial fibrillary acid protein (GFAP), enteric glial marker, and GPx protein content and activity were assessed in the colon. The major phyla of gut microbiota were assessed.Results: CKD-BN group presented a decrease in GFAP content (p = 0.0001). The CKD-BN group modulated the abundance of Firmicutes, increasing its proportion compared to the CKD group. The CKD-BN group showed increased GPx activity in the colon (p = 0.0192), despite no significant difference in protein content.Conclusion: Brazil nut-enriched diet consumption decreased enteric glial reactivity and modulated gut microbiota in the CKD experimental model.
Collapse
Affiliation(s)
- Patricia Pereira Almeida
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Beatriz Oliveira Da Cruz
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Beatriz Thomasi
- Department of Physiology, Neuroscience Program, Michigan State University (MSU), East Lansing, Michigan, USA
| | - Ágatha Cristie Menezes
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Michele Lima Brito
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Nathalia da Silva Costa
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | | | - Viviane Alexandre Nunes Degani
- Clinic and Animal Reproduction Post Graduate Program, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Julio Beltrame Daleprane
- Laboratory for Studies of Interactions Between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - D'Angelo Carlo Magliano
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Ana Lúcia Tavares-Gomes
- Neurosciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Milena Barcza Stockler-Pinto
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
- Nutrition Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| |
Collapse
|
71
|
Álvarez-Herms J, González-Benito A, Corbi F, Odriozola A. What if gastrointestinal complications in endurance athletes were gut injuries in response to a high consumption of ultra-processed foods? Please take care of your bugs if you want to improve endurance performance: a narrative review. Eur J Appl Physiol 2024; 124:383-402. [PMID: 37839038 DOI: 10.1007/s00421-023-05331-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 09/20/2023] [Indexed: 10/17/2023]
Abstract
To improve performance and recovery faster, athletes are advised to eat more often than usual and consume higher doses of simple carbohydrates, during and after exercise. Sports energetic supplements contain food additives, such as artificial sweeteners, emulsifiers, acidity regulators, preservatives, and salts, which could be harmful to the gut microbiota and impair the intestinal barrier function. The intestinal barrier plays a critical function in bidirectionally regulation of the selective transfer of nutrients, water, and electrolytes, while preventing at the same time, the entrance of harmful substances (selective permeability). The gut microbiota helps to the host to regulate intestinal homeostasis through metabolic, protective, and immune functions. Globally, the gut health is essential to maintain systemic homeostasis in athletes, and to ensure proper digestion, metabolization, and substrate absorption. Gastrointestinal complaints are an important cause of underperformance and dropout during endurance events. These complications are directly related to the loss of gut equilibrium, mainly linked to microbiota dysbiosis and leaky gut. In summary, athletes must be cautious with the elevated intake of ultra-processed foods and specifically those contained on sports nutrition supplements. This review points out the specific nutritional interventions that should be implemented and/or discontinued depending on individual gut functionality.
Collapse
Affiliation(s)
- Jesús Álvarez-Herms
- Phymolab (Physiology and Molecular Laboratory), Collado Hermoso, Segovia, Spain.
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Bilbao, Spain.
| | - A González-Benito
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - F Corbi
- Institut Nacional d'Educació Física de Catalunya (INEFC), University of Lleida (UdL), Lleida, Spain
| | - A Odriozola
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| |
Collapse
|
72
|
Pei W, Li M, Wu J, Huang M, Sun B, Liang H, Wu Z. Preparation, Structural Analysis, and Intestinal Probiotic Properties of a Novel Oligosaccharide from Enzymatic Degradation of Huangshui Polysaccharide. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:313-325. [PMID: 38126348 DOI: 10.1021/acs.jafc.3c05666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Huangshui polysaccharide (HSP) has attracted more and more interest due to its potential health benefits. Despite being an excellent source for the preparation of oligosaccharides, there are currently no relevant research reports on HSP. In the present study, a novel oligosaccharide (HSO) with a molecular weight of 1791 Da and a degree of polymerization of 11 was prepared through enzymatic degradation of crude HSP (cHSP). Methylation and NMR analyses revealed that the main chain of HSO was (1 → 4)-α-d-glucose with two O-6-linked branched chains. Morphological observations indicated that HSO exhibited smooth surface with lamellar and filamentary structure, and the glycan size ranged from 0.03 to 0.20 μm. Notably, HSO significantly promoted the proliferation of Bifidobacterium, Bacteroides, and Phascolarctobacterium, thereby making positive alterations in intestinal microbiota composition. Moreover, HSO markedly increased the content of short-chain fatty acids during in vitro fermentation. Metabolomics analysis illustrated the important metabolic pathways primarily involving glucose metabolism, amino acid metabolism, and fatty acid metabolism.
Collapse
Affiliation(s)
- Wenhao Pei
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, China
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing 100048, China
| | - Mei Li
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, China
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing 100048, China
| | - Jihong Wu
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, China
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing 100048, China
| | - Mingquan Huang
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, China
| | - Baoguo Sun
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, China
| | - Haiyan Liang
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing 100048, China
| | - Ziyan Wu
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, China
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
73
|
Zhou R, Huang Y, Feng X, Zhou R, Wang L, Xie G, Xiao Y, Zhou H. Decreased YB-1 expression denervates brown adipose tissue and contributes to age-related metabolic dysfunction. Cell Prolif 2024; 57:e13520. [PMID: 37321837 PMCID: PMC10771110 DOI: 10.1111/cpr.13520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/17/2023] Open
Abstract
Thermogenesis in brown adipose tissue (BAT) declines with aging, however, the underlying mechanism remains unclear. Here, we show that the expression of Y-box binding protein 1 (YB-1), a critical DNA/RNA binding protein, decreased in the BAT of aged mice due to the reduction of microbial metabolite butyrate. Genetic ablation of YB-1 in the BAT accelerated diet-induced obesity and BAT thermogenic dysfunction. In contrast, overexpression of YB-1 in the BAT of aged mice was sufficient to promote BAT thermogenesis, thus alleviating diet-induced obesity and insulin resistance. Interestingly, YB-1 had no direct effect on adipose UCP1 expression. Instead, YB-1 promoted axon guidance of BAT via regulating the expression of Slit2, thus potentiating sympathetic innervation and thermogenesis. Moreover, we have identified that a natural compound Sciadopitysin, which promotes YB-1 protein stability and nuclear translocation, alleviated BAT aging and metabolic disorders. Together, we reveal a novel fat-sympathetic nerve unit in regulating BAT senescence and provide a promising strategy against age-related metabolic disorders.
Collapse
Affiliation(s)
- Ruoyu Zhou
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Xu Feng
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Rui Zhou
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Liwen Wang
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Genqing Xie
- Department of EndocrinologyThe First People's Hospital of Xiangtan cityXiangtanChina
| | - Yuan Xiao
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalChangshaChina
| | - Haiyan Zhou
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalChangshaChina
| |
Collapse
|
74
|
Liu X, Tang H, Huang X, Xu M. Butyrate affects bacterial virulence: a new perspective on preventing enteric bacterial pathogen invasion. Future Microbiol 2024; 19:73-84. [PMID: 38085176 DOI: 10.2217/fmb-2023-0148] [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: 06/29/2023] [Accepted: 09/11/2023] [Indexed: 02/15/2024] Open
Abstract
Enteric bacterial pathogens are a major threat to intestinal health. With the widespread use of antibiotics, bacterial resistance has become a problem, and there is an urgent need for a new treatment to reduce dependence on antibiotics. Butyrate can control enteric bacterial pathogens by regulating the expression of their virulence genes, promoting the posttranslational modification of their proteins, maintaining an anaerobic environment, regulating the host immune system and strengthening the intestinal mucosal barrier. Here, this review describes the mechanisms by which butyrate regulates the pathogenicity of enteric bacterial pathogens from various perspectives and discusses the prospects and limitations of butyrate as a new option for the control of pathogenic bacteria.
Collapse
Affiliation(s)
- Xiucheng Liu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212008, China
- Department of Biochemistry & Molecular Biology, Jiangsu University School of Medicine, Zhenjiang, Jiangsu, 212013, China
| | - Hao Tang
- Department of Biochemistry & Molecular Biology, Jiangsu University School of Medicine, Zhenjiang, Jiangsu, 212013, China
| | - Xinxiang Huang
- Department of Biochemistry & Molecular Biology, Jiangsu University School of Medicine, Zhenjiang, Jiangsu, 212013, China
| | - Min Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212008, China
- Institute of Digestive Diseases, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| |
Collapse
|
75
|
Cruz MS, Tintelnot J, Gagliani N. Roles of microbiota in pancreatic cancer development and treatment. Gut Microbes 2024; 16:2320280. [PMID: 38411395 PMCID: PMC10900280 DOI: 10.1080/19490976.2024.2320280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/14/2024] [Indexed: 02/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with poor prognosis. This is due to the fact that most cases are only diagnosed at an advanced and palliative disease stage, and there is a high incidence of therapy resistance. Despite ongoing efforts, to date, the mechanisms underlying PDAC oncogenesis and its poor responses to treatment are still largely unclear. As the study of the microbiome in cancer progresses, growing evidence suggests that bacteria or fungi might be key players both in PDAC oncogenesis as well as in its resistance to chemo- and immunotherapy, for instance through modulation of the tumor microenvironment and reshaping of the host immune response. Here, we review how the microbiota exerts these effects directly or indirectly via microbial-derived metabolites. Finally, we further discuss the potential of modulating the microbiota composition as a therapy in PDAC.
Collapse
Affiliation(s)
- Mariana Santos Cruz
- II. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), Hamburg, Germany
| | - Joseph Tintelnot
- II. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), Hamburg, Germany
| | - Nicola Gagliani
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), Hamburg, Germany
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
76
|
Hishiya N, Uno K, Nakano A, Konishi M, Higashi S, Eguchi S, Ariyoshi T, Matsumoto A, Oka K, Takahashi M, Suzuki Y, Horiuchi S, Hirai N, Ogawa Y, Ogawa T, Nakano R, Mikasa K, Kasahara K, Yano H. Association between the gut microbiome and organic acid profiles in a Japanese population with HIV infection. J Infect Chemother 2024; 30:58-66. [PMID: 37708940 DOI: 10.1016/j.jiac.2023.09.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 09/16/2023]
Abstract
INTRODUCTION An increased incidence of metabolic syndrome has been observed in human immunodeficiency virus (HIV)-infected individuals. In contrast, gut dysbiosis is involved in various pathogeneses, including vascular endothelial disorders. Organic acids, including short-chain fatty acids (SCFAs), are essential for maintaining gut homeostasis. Therefore, this study aimed to explore the gut microbiome profile and organic acids in a Japanese population infected with HIV. METHODS Forty-nine patients with HIV infection on combination antiretroviral therapy (cART) were enrolled and divided into the high and low CD4 groups based on a CD4 cutoff of 350 cells/μL. Stool samples were analyzed by 16S ribosomal RNA next-generation sequencing and high-performance liquid chromatography. The association between the gut microbiome, including bacterial taxa and organic acids, was statistically analyzed. RESULTS The fecal microbial community composition was significantly different between HIV patients with CD4 counts above and below 350 cells/μL. The relative abundance of Roseburia, Prevotella, Prevotella_9, and [Clostridium]_methylpentosum_group were significantly enriched in the high CD4 group. Fecal succinic acid tended to be more abundant in the low CD4 group, and acetic, propionic, and butyric acids tended to be more abundant in the high CD4 group. Roseburia was positively correlated with butyric acid levels. Prevotella_9 and Prevotella were negatively correlated with succinic acid levels and positively correlated with acetic and propionic acid levels. CONCLUSIONS This study showed intestinal dysbiosis bordering on a CD4 count of 350 in patients with HIV infection undergoing cART. These findings might help in understanding intestinal damage and systemic inflammation in HIV infection.
Collapse
Affiliation(s)
- Naokuni Hishiya
- Department of Microbiology and Infectious Diseases, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8521, Japan; Department of Infectious Diseases, Nara City Hospital, 1-50-1 Higashikidera-cho, Nara-Shi, Nara, 630-8305, Japan
| | - Kenji Uno
- Department of Infectious Diseases, Minami-Nara General Medical Center, 8-1 Fukugami, Oyodo-Cho, Yoshino-Gun, Nara, 638-8551, Japan
| | - Akiyo Nakano
- Department of Microbiology and Infectious Diseases, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8521, Japan.
| | - Mitsuru Konishi
- Center for Health Control, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8521, Japan; Center for Infectious Diseases, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8521, Japan
| | - Seiya Higashi
- R&D Division, Miyarisan Pharmaceutical Co., Ltd., 2-22-9 Toro-Cho, Kita-Ku, Saitama-Shi, Saitama, 331-0804, Japan
| | - Shuhei Eguchi
- R&D Division, Miyarisan Pharmaceutical Co., Ltd., 2-22-9 Toro-Cho, Kita-Ku, Saitama-Shi, Saitama, 331-0804, Japan
| | - Tadashi Ariyoshi
- R&D Division, Miyarisan Pharmaceutical Co., Ltd., 2-22-9 Toro-Cho, Kita-Ku, Saitama-Shi, Saitama, 331-0804, Japan
| | - Asami Matsumoto
- R&D Division, Miyarisan Pharmaceutical Co., Ltd., 2-22-9 Toro-Cho, Kita-Ku, Saitama-Shi, Saitama, 331-0804, Japan
| | - Kentaro Oka
- R&D Division, Miyarisan Pharmaceutical Co., Ltd., 2-22-9 Toro-Cho, Kita-Ku, Saitama-Shi, Saitama, 331-0804, Japan
| | - Motomichi Takahashi
- R&D Division, Miyarisan Pharmaceutical Co., Ltd., 2-22-9 Toro-Cho, Kita-Ku, Saitama-Shi, Saitama, 331-0804, Japan
| | - Yuki Suzuki
- Department of Microbiology and Infectious Diseases, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8521, Japan
| | - Saori Horiuchi
- Department of Microbiology and Infectious Diseases, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8521, Japan
| | - Nobuyasu Hirai
- Department of Microbiology and Infectious Diseases, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8521, Japan; Department of Gastroenterology, Seichokai Fuchu Hospital, 1-10-17 Hiko-Cho, Izumi, Osaka, 594-0076, Japan
| | - Yoshihiko Ogawa
- Department of Infectious Diseases, Sakai City Medical Center, 1-1-1 Ebaraji-Cho, Nishi-Ku, Sakai, Osaka, 593-8304, Japan
| | - Taku Ogawa
- Department of Microbiology and Infection Control, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki, Osaka, 569-8686, Japan
| | - Ryuichi Nakano
- Department of Microbiology and Infectious Diseases, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8521, Japan
| | - Keiichi Mikasa
- Center for Infectious Diseases, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8521, Japan; Department of Internal Medicine, Nara Koseikai Hospital, 769-3 Shigi-cho, Yamatokoriyama, Nara, 639-1039, Japan
| | - Kei Kasahara
- Center for Infectious Diseases, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8521, Japan
| | - Hisakazu Yano
- Department of Microbiology and Infectious Diseases, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8521, Japan
| |
Collapse
|
77
|
Xiao L, Zhang C, Zhang X, Zhao X, Chaeipeima Mahsa G, Ma K, Ji F, Azarpazhooh E, Ajami M, Rui X, Li W. Effects of Lacticaseibacillus paracasei SNB-derived postbiotic components on intestinal barrier dysfunction and composition of gut microbiota. Food Res Int 2024; 175:113773. [PMID: 38129062 DOI: 10.1016/j.foodres.2023.113773] [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: 08/13/2023] [Revised: 11/09/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023]
Abstract
The bacterial surface components are considered as effector molecules and show the potential to support intestinal health, but the detailed mechanism of how the gut microbiota changes after the intervention of surface molecules is still unknown. In the present study, capsular polysaccharide (B-CPS) and surface layer protein (B-SLP) were extracted from Lacticaseibacillus paracasei S-NB. The protective effect of direct administration of B-CPS (100 μg/mL) and B-SLP (100 μg/mL) on intestinal epithelial barrier dysfunction was verified based on the LPS-induced Caco-2 cell model. Additionally, the B-CPS and B-SLP could be utilized as carbon source and nitrogen source for the growth of several Lactobacillus strains, respectively. The postbiotic potential of B-CPS and B-SLP was further evaluated by in vitro fermentation with fecal cultures. The B-CPS and a combination of B-CPS and B-SLP regulated the composition of gut microbiota by increasing the relative abundances of Bacteroides, Bifidobacterium, Phascolarctobacterium, Parabacteroides, Subdoligranulum and Collinsella and decreasing the abundance of pathogenic bacteria like Escherichia-Shigella, Blautia, Citrobacter and Fusobacterium. Meanwhile, the total short-chain fatty acid production markedly increased after fermentation with either B-CPS individually or in combination with B-SLP. These results provided an important basis for the application of B-CPS and B-SLP as postbiotics to improve human intestinal health.
Collapse
Affiliation(s)
- Luyao Xiao
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Changliang Zhang
- Jiangsu New-Bio Biotechnology Co., Ltd, Jiangyin, Jiangsu 214400, PR China; Jiangsu Biodep Biotechnology Co., Ltd, Jiangyin, Jiangsu 214400, PR China
| | - Xueliang Zhang
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Xiaogan Zhao
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Ghahvechi Chaeipeima Mahsa
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Kai Ma
- Jiangsu New-Bio Biotechnology Co., Ltd, Jiangyin, Jiangsu 214400, PR China; Jiangsu Biodep Biotechnology Co., Ltd, Jiangyin, Jiangsu 214400, PR China
| | - Feng Ji
- Jiangsu New-Bio Biotechnology Co., Ltd, Jiangyin, Jiangsu 214400, PR China; Jiangsu Biodep Biotechnology Co., Ltd, Jiangyin, Jiangsu 214400, PR China
| | - Elham Azarpazhooh
- Khorasan Razavi Agricultural and Natural Resources Research and Education Center, AREEO, Iran
| | - Marjan Ajami
- National Nutrition and Food Technology Research Institute, School of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Xin Rui
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Wei Li
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China.
| |
Collapse
|
78
|
Liu L, Kaur GI, Kumar A, Kanwal A, Singh SP. The Role of Gut Microbiota and Associated Compounds in Cardiovascular Health and its Therapeutic Implications. Cardiovasc Hematol Agents Med Chem 2024; 22:375-389. [PMID: 38275032 DOI: 10.2174/0118715257273506231208045308] [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: 08/05/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 01/27/2024]
Abstract
It is possible that gut bacteria may have a beneficial effect on cardiovascular health in humans. It may play a major role in the progression of a variety of cardiovascular diseases, including Heart Failure (HF), Atherosclerosis, Coronary Arterial Disease (CAD), Ischemic Heart Disease (IHD), and Others. Dysbiosis of the gut microbiota, along with its direct and indirect impact on gut health, may induce cardiovascular disorders. Although advanced studies have demonstrated the relationship of various metabolites to cardiovascular diseases (CVD) in animals, translating their functional capacity to humans remains a significant area of research. This paper simplifies the demonstration of some compounds, pathways, and components like Trimethylamine N-oxide (TMAO), short-chain fatty acids (SCFAs), and butyrate production. It demonstrates how a change in eating habits causes TMAO and how the impact of different drugs on gut microbiota species and high consumption of Westernized food causes several heartrelated problems, such as atherosclerosis and inflammation that can even become the cause of heart failure. Modulation of the gut microbiome, on the other hand, is a novel therapeutic measure because it can be easily altered through diet and other lifestyle changes. It could then be used to lower the risk of several CVDs.
Collapse
Affiliation(s)
- Lu Liu
- Endoscopic Diagnosis and Treatment Center, Baoding First Central Hospital, Baoding, China
| | - Guneet Inderjeet Kaur
- Department of Sports Psychology, Central University of Rajasthan, Ajmer, 305817, India
| | - Avinash Kumar
- Department of Sports Biosciences, Central University of Rajasthan, Ajmer, 305817, India
| | | | | |
Collapse
|
79
|
Xu X, Xu T, Wei J, Chen T. Gut microbiota: an ideal biomarker and intervention strategy for aging. MICROBIOME RESEARCH REPORTS 2024; 3:13. [PMID: 38841415 PMCID: PMC11149087 DOI: 10.20517/mrr.2023.68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/15/2023] [Indexed: 06/07/2024]
Abstract
Population aging is a substantial challenge for the global sanitation framework. Unhealthy aging tends to be accompanied by chronic diseases such as cardiovascular disease, diabetes, and cancer, which undermine the welfare of the elderly. Based on the fact that aging is inevitable but retarding aging is attainable, flexible aging characterization and efficient anti-aging become imperative for healthy aging. The gut microbiome, as the most dynamic component interacting with the organism, can affect the aging process through its own structure and metabolites, thus holding the potential to become both an ideal aging-related biomarker and an intervention strategy. This review summarizes the value of applying gut microbiota as aging-related microbial biomarkers in diagnosing aging state and monitoring the effect of anti-aging interventions, ultimately pointing to the future prospects of microbial intervention strategies in maintaining healthy aging.
Collapse
Affiliation(s)
- Xuan Xu
- Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
- Huankui Academy, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
- Authors contributed equally
| | - Tangchang Xu
- Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
- Authors contributed equally
| | - Jing Wei
- Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Tingtao Chen
- Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
| |
Collapse
|
80
|
Ambat A, Antony L, Maji A, Ghimire S, Mattiello S, Kashyap PC, More S, Sebastian V, Scaria J. Enhancing recovery from gut microbiome dysbiosis and alleviating DSS-induced colitis in mice with a consortium of rare short-chain fatty acid-producing bacteria. Gut Microbes 2024; 16:2382324. [PMID: 39069899 PMCID: PMC11290756 DOI: 10.1080/19490976.2024.2382324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 05/24/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024] Open
Abstract
The human gut microbiota is a complex community comprising hundreds of species, with a few present in high abundance and the vast majority in low abundance. The biological functions and effects of these low-abundant species on their hosts are not yet fully understood. In this study, we assembled a bacterial consortium (SC-4) consisting of B. paravirosa, C. comes, M. indica, and A. butyriciproducens, which are low-abundant, short-chain fatty acid (SCFA)-producing bacteria isolated from healthy human gut, and tested its effect on host health using germ-free and human microbiota-associated colitis mouse models. The selection also favored these four bacteria being reduced in abundance in either Ulcerative Colitis (UC) or Crohn's disease (CD) metagenome samples. Our findings demonstrate that SC-4 can colonize germ-free (GF) mice, increasing mucin thickness by activating MUC-1 and MUC-2 genes, thereby protecting GF mice from Dextran Sodium Sulfate (DSS)-induced colitis. Moreover, SC-4 aided in the recovery of human microbiota-associated mice from DSS-induced colitis, and intriguingly, its administration enhanced the alpha diversity of the gut microbiome, shifting the community composition closer to control levels. The results showed enhanced phenotypes across all measures when the mice were supplemented with inulin as a dietary fiber source alongside SC-4 administration. We also showed a functional redundancy existing in the gut microbiome, resulting in the low abundant SCFA producers acting as a form of insurance, which in turn accelerates recovery from the dysbiotic state upon the administration of SC-4. SC-4 colonization also upregulated iNOS gene expression, further supporting its ability to produce an increasing number of goblet cells. Collectively, our results provide evidence that low-abundant SCFA-producing species in the gut may offer a novel therapeutic approach to IBD.
Collapse
Affiliation(s)
- Achuthan Ambat
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Linto Antony
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA
| | - Abhijit Maji
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA
| | - Sudeep Ghimire
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA
| | - Samara Mattiello
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA
| | - Purna C. Kashyap
- Enteric Neuroscience Program, Department of Medicine and Physiology, Mayo Clinic, Rochester, MN, USA
| | - Sunil More
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Vanessa Sebastian
- Department of Pathology, Jubilee Mission Medical College and Research Institute, Thrissur, India
| | - Joy Scaria
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| |
Collapse
|
81
|
Xu X, Zhao Y, Wang X, Zhang R, Liu S, Sun R, Wang Z, Ge L, Sun Y, Zhang S, Ma H, Zhan J. Diagnostic and prognostic value of the gut microbiota and its metabolite butyrate in children with biliary atresia. Pediatr Surg Int 2023; 40:24. [PMID: 38127131 DOI: 10.1007/s00383-023-05606-8] [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] [Accepted: 11/21/2023] [Indexed: 12/23/2023]
Abstract
PURPOSE To determine the prevalent microbiological profile of biliary atresia (BA) patients at the time of its occurrence by studying their intestinal flora. METHODS A total of 118 gut microbiota samples from three groups of 43 BA patients, 33 disease controls (DC) with other cholestatic diseases and 42 healthy controls (HC), were analyzed by deep mining of public data. Subsequently, a total of 23 fecal samples from three groups of clinically collected patients (11 BA, 6 DC and 6 HC) were sequenced for 16S rRNA gene amplification and analyzed for serum butyrate (BU) level by liquid chromatography. RESULTS Taxonomic analysis revealed significant differences in the composition of the intestinal microbiota between BA patients and controls, with a reduction in diversity and a higher abundance of Proteobacteria, Streptococcus and Lactobacillus in the BA group. Database and clinical data analyses concluded that Streptococcus/Bacteroides (AUC = 0.9035, 95% CI 0.8347-0.9722, P < 0.0001) or Streptococcus/Eggerthella (AUC = 0.8333, 95% CI 0.6340-1.000, P = 0.027) was the best microbiota to differentiate between BA and DC. Serum butyrate levels were low in the BA and DC groups and differed from the HC group (P = 0.01, P = 0.04). Butyrate levels in BA were negatively correlated with jaundice clearance and cholangitis, but not statistically significant. CONCLUSIONS Our study reveals changes in the composition of the gut microbiota in BA, especially the butyrate-producing microbiota, and suggests the potential for using gut microbiota as a noninvasive diagnostic benefit for BA. Low levels of serum butyrate in BA may indicate a poor prognosis.
Collapse
Affiliation(s)
- Xiaodan Xu
- Graduate College, Tianjin Medical University, Tianjin, 300070, People's Republic of China
- Department of General Surgery, Tianjin Children's Hospital, LongYan Road 238, Beichen District, Tianjin, 300134, People's Republic of China
| | - Yilin Zhao
- Graduate College, Tianjin Medical University, Tianjin, 300070, People's Republic of China
- Department of General Surgery, Tianjin Children's Hospital, LongYan Road 238, Beichen District, Tianjin, 300134, People's Republic of China
| | - Xueting Wang
- Graduate College, Tianjin Medical University, Tianjin, 300070, People's Republic of China
- Department of Pediatric Surgery, Xinjiang Yili Friendship Hospital, Yili, 835000, People's Republic of China
| | - Ruifeng Zhang
- Graduate College, Tianjin Medical University, Tianjin, 300070, People's Republic of China
- Department of General Surgery, Tianjin Children's Hospital, LongYan Road 238, Beichen District, Tianjin, 300134, People's Republic of China
| | - Shaowen Liu
- Graduate College, Tianjin Medical University, Tianjin, 300070, People's Republic of China
- Department of General Surgery, Tianjin Children's Hospital, LongYan Road 238, Beichen District, Tianjin, 300134, People's Republic of China
| | - Rongjuan Sun
- Graduate College, Tianjin Medical University, Tianjin, 300070, People's Republic of China
- Department of General Surgery, Tianjin Children's Hospital, LongYan Road 238, Beichen District, Tianjin, 300134, People's Republic of China
| | - Zhiru Wang
- Graduate College, Tianjin Medical University, Tianjin, 300070, People's Republic of China
- Department of General Surgery, Tianjin Children's Hospital, LongYan Road 238, Beichen District, Tianjin, 300134, People's Republic of China
| | - Liang Ge
- Department of General Surgery, Tianjin Children's Hospital, LongYan Road 238, Beichen District, Tianjin, 300134, People's Republic of China
| | - Yan Sun
- Department of General Surgery, Tianjin Children's Hospital, LongYan Road 238, Beichen District, Tianjin, 300134, People's Republic of China
| | - Shujian Zhang
- Department of General Surgery, Tianjin Children's Hospital, LongYan Road 238, Beichen District, Tianjin, 300134, People's Republic of China
| | - Hui Ma
- Department of Clinical Laboratory, Tianjin Children's Hospital, Tianjin, 300134, People's Republic of China
| | - Jianghua Zhan
- Department of General Surgery, Tianjin Children's Hospital, LongYan Road 238, Beichen District, Tianjin, 300134, People's Republic of China.
| |
Collapse
|
82
|
Alghetaa H, Mohammed A, Singh NP, Bloomquist RF, Chatzistamou I, Nagarkatti M, Nagarkatti P. Estrobolome dysregulation is associated with altered immunometabolism in a mouse model of endometriosis. Front Endocrinol (Lausanne) 2023; 14:1261781. [PMID: 38144564 PMCID: PMC10748389 DOI: 10.3389/fendo.2023.1261781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 11/17/2023] [Indexed: 12/26/2023] Open
Abstract
Introduction Endometriosis is a painful disease that affects around 5% of women of reproductive age. In endometriosis, ectopic endometrial cells or seeded endometrial debris grow in abnormal locations including the peritoneal cavity. Common manifestations of endometriosis include dyspareunia, dysmenorrhea, chronic pelvic pain and often infertility and symptomatic relief or surgical removal are mainstays of treatment. Endometriosis both promotes and responds to estrogen imbalance, leading to intestinal bacterial estrobolome dysregulation and a subsequent induction of inflammation. Methods In the current study, we investigated the linkage between gut dysbiosis and immune metabolic response in endometriotic mice. Ovariectomized BALB/c mice received intraperitoneal transplantation of endometrial tissue from OVX donors (OVX+END). Control groups included naïve mice (Naïve), naïve mice that received endometrial transplants (Naive+END) and OVX mice that received the vehicle (OVX+VEH). Colonic content was collected 2 weeks post-transplantation for 16s rRNA pyrosequencing and peritoneal fluid was collected to determine the phenotype of inflammatory cells by flow cytometry. Results We noted a significant increase in the number of peritoneal fluid cells, specifically, T cells, natural killer (NK) cells, and NKT cells in OVX+END mice. Phylogenetic taxonomy analysis showed significant dysbiosis in OVX+END mice, with an increase in abundance of Phylum Tenericutes, Class Mollicutes, Order Aneroplasmatales, and Genus Aneroplasma, and a decrease in Order Clostridiales, and Genus Dehalobacterium, when compared to OVX+VEH controls. The metabolomic profile showed an increase in some tricarboxylic acid cycle (TCA)-related metabolites accompanied by a reduction in short-chain fatty acids (SCFA) such as butyric acid in OVX+END mice. Additionally, the mitochondrial and ATP production of immune cells was enforced to a maximal rate in OVX+END mice when compared to OVX+VEH mice. Conclusion The current study demonstrates that endometriosis alters the gut microbiota and associated immune metabolism.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
83
|
Yan M, Man S, Sun B, Ma L, Guo L, Huang L, Gao W. Gut liver brain axis in diseases: the implications for therapeutic interventions. Signal Transduct Target Ther 2023; 8:443. [PMID: 38057297 PMCID: PMC10700720 DOI: 10.1038/s41392-023-01673-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/10/2023] [Accepted: 09/28/2023] [Indexed: 12/08/2023] Open
Abstract
Gut-liver-brain axis is a three-way highway of information interaction system among the gastrointestinal tract, liver, and nervous systems. In the past few decades, breakthrough progress has been made in the gut liver brain axis, mainly through understanding its formation mechanism and increasing treatment strategies. In this review, we discuss various complex networks including barrier permeability, gut hormones, gut microbial metabolites, vagus nerve, neurotransmitters, immunity, brain toxic metabolites, β-amyloid (Aβ) metabolism, and epigenetic regulation in the gut-liver-brain axis. Some therapies containing antibiotics, probiotics, prebiotics, synbiotics, fecal microbiota transplantation (FMT), polyphenols, low FODMAP diet and nanotechnology application regulate the gut liver brain axis. Besides, some special treatments targeting gut-liver axis include farnesoid X receptor (FXR) agonists, takeda G protein-coupled receptor 5 (TGR5) agonists, glucagon-like peptide-1 (GLP-1) receptor antagonists and fibroblast growth factor 19 (FGF19) analogs. Targeting gut-brain axis embraces cognitive behavioral therapy (CBT), antidepressants and tryptophan metabolism-related therapies. Targeting liver-brain axis contains epigenetic regulation and Aβ metabolism-related therapies. In the future, a better understanding of gut-liver-brain axis interactions will promote the development of novel preventative strategies and the discovery of precise therapeutic targets in multiple diseases.
Collapse
Affiliation(s)
- Mengyao Yan
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China
| | - Shuli Man
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China.
| | - Benyue Sun
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China
| | - Long Ma
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China
| | - Lanping Guo
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700, Beijing, China.
| | - Luqi Huang
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, 300072, Tianjin, China.
| |
Collapse
|
84
|
Lemaigre S, Gerin PA, Adam G, Klimek D, Goux X, Herold M, Frkova Z, Calusinska M, Delfosse P. Potential of acetic acid to restore methane production in anaerobic reactors critically intoxicated by ammonia as evidenced by metabolic and microbial monitoring. BIOTECHNOLOGY FOR BIOFUELS AND BIOPRODUCTS 2023; 16:188. [PMID: 38042839 PMCID: PMC10693713 DOI: 10.1186/s13068-023-02438-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 11/21/2023] [Indexed: 12/04/2023]
Abstract
BACKGROUND Biogas and biomethane production from the on-farm anaerobic digestion (AD) of animal manure and agri-food wastes could play a key role in transforming Europe's energy system by mitigating its dependence on fossil fuels and tackling the climate crisis. Although ammonia is essential for microbial growth, it inhibits the AD process if present in high concentrations, especially under its free form, thus leading to economic losses. In this study, which includes both metabolic and microbial monitoring, we tested a strategy to restore substrate conversion to methane in AD reactors facing critical free ammonia intoxication. RESULTS The AD process of three mesophilic semi-continuous 100L reactors critically intoxicated by free ammonia (> 3.5 g_N L-1; inhibited hydrolysis and heterotrophic acetogenesis; interrupted methanogenesis) was restored by applying a strategy that included reducing pH using acetic acid, washing out total ammonia with water, re-inoculation with active microbial flora and progressively re-introducing sugar beet pulp as a feed substrate. After 5 weeks, two reactors restarted to hydrolyse the pulp and produced CH4 from the methylotrophic methanogenesis pathway. The acetoclastic pathway remained inhibited due to the transient dominance of a strictly methylotrophic methanogen (Candidatus Methanoplasma genus) to the detriment of Methanosarcina. Concomitantly, the third reactor, in which Methanosarcina remained dominant, produced CH4 from the acetoclastic pathway but faced hydrolysis inhibition. After 11 weeks, the hydrolysis, the acetoclastic pathway and possibly the hydrogenotrophic pathway were functional in all reactors. The methylotrophic pathway was no longer favoured. Although syntrophic propionate oxidation remained suboptimal, the final pulp to CH4 conversion ratio (0.41 ± 0.10 LN_CH4 g_VS-1) was analogous to the pulp biochemical methane potential (0.38 ± 0.03 LN_CH4 g_VS-1). CONCLUSIONS Despite an extreme free ammonia intoxication, the proposed process recovery strategy allowed CH4 production to be restored in three intoxicated reactors within 8 weeks, a period during which re-inoculation appeared to be crucial to sustain the process. Introducing acetic acid allowed substantial CH4 production during the recovery period. Furthermore, the initial pH reduction promoted ammonium capture in the slurry, which could allow the field application of the effluents produced by full-scale digesters recovering from ammonia intoxication.
Collapse
Affiliation(s)
- Sébastien Lemaigre
- Environmental Research and Innovation Department, Luxembourg Institute of Science and Technology, Rue du Brill 41, L-4422, Belvaux, Luxembourg.
| | - Patrick A Gerin
- Earth and Life Institute, Bioengineering, Université Catholique de Louvain, Croix du Sud 2, Box L7.05.19, B-1348, Louvain-la-Neuve, Belgium
| | - Gilles Adam
- Environmental Research and Innovation Department, Luxembourg Institute of Science and Technology, Rue du Brill 41, L-4422, Belvaux, Luxembourg
| | - Dominika Klimek
- Environmental Research and Innovation Department, Luxembourg Institute of Science and Technology, Rue du Brill 41, L-4422, Belvaux, Luxembourg
| | - Xavier Goux
- Environmental Research and Innovation Department, Luxembourg Institute of Science and Technology, Rue du Brill 41, L-4422, Belvaux, Luxembourg
| | - Malte Herold
- Environmental Research and Innovation Department, Luxembourg Institute of Science and Technology, Rue du Brill 41, L-4422, Belvaux, Luxembourg
| | - Zuzana Frkova
- Environmental Research and Innovation Department, Luxembourg Institute of Science and Technology, Rue du Brill 41, L-4422, Belvaux, Luxembourg
| | - Magdalena Calusinska
- Environmental Research and Innovation Department, Luxembourg Institute of Science and Technology, Rue du Brill 41, L-4422, Belvaux, Luxembourg
| | - Philippe Delfosse
- Université du Luxembourg, Campus Belval, Maison du Savoir, Avenue de l'Université 2, L-4365, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
85
|
Xia H, Guo J, Shen J, Jiang S, Han S, Li L. Butyrate ameliorated the intestinal barrier dysfunction and attenuated acute pancreatitis in mice fed with ketogenic diet. Life Sci 2023; 334:122188. [PMID: 37866809 DOI: 10.1016/j.lfs.2023.122188] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/07/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023]
Abstract
Butyrate, a short-chain fatty acid (SCFA), has demonstrated significant efficacy in preventing colitis-associated inflammation. Acute pancreatitis is an acute gastrointestinal disorder characterized by increased systemic inflammation, bacterial translocation, and disrupted intestinal barrier. However, the effects and mechanisms of butyrate in attenuating acute pancreatitis remain unclear. In this study, we established two mouse models of acute pancreatitis induced by cerulein (Cer) and taurocholate (TA), which were further exacerbated by a ketogenic diet (KD). The results suggested that butyrate supplementation effectively reduced mortality rates, systemic inflammation, and intestinal barrier disruption caused by Cer- and TA-induced acute pancreatitis in mice fed a KD. Furthermore, we observed a significant reduction in gut microbiota diversity as well as overgrowth of Lachnospirales and Erysipelotrichales along with depletion of SCFAs in mice fed a KD, and these alterations were reversed by butyrate supplement. To evaluate the role of microbiota and butyrate supplement, we conducted germ-depletion trials by antibiotics. The results showed that while systemic inflammation was attenuated in mice with TA-induced pancreatitis following antibiotic treatment, the reduction in mortality remained inconclusive (p = 0.055). Importantly, the key differential change between antibiotic treatment and butyrate supplementation was found to be related to intestinal barrier dysfunction and repairment. These results suggest that butyrate plays a central role in mitigating acute pancreatitis through amelioration of intestinal barrier dysfunction.
Collapse
Affiliation(s)
- He Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine,79 Qingchun Rd., Hangzhou City 310003, China
| | - Jing Guo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine,79 Qingchun Rd., Hangzhou City 310003, China
| | - Jian Shen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine,79 Qingchun Rd., Hangzhou City 310003, China
| | - Shiman Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine,79 Qingchun Rd., Hangzhou City 310003, China
| | - Shengyi Han
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine,79 Qingchun Rd., Hangzhou City 310003, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine,79 Qingchun Rd., Hangzhou City 310003, China.
| |
Collapse
|
86
|
Tortadès M, Marti S, Devant M, Vidal M, Fàbregas F, Terré M. Feeding colostrum and transition milk facilitates digestive tract functionality recovery from feed restriction and fasting of dairy calves. J Dairy Sci 2023; 106:8642-8657. [PMID: 37641341 DOI: 10.3168/jds.2023-23345] [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: 02/06/2023] [Accepted: 06/14/2023] [Indexed: 08/31/2023]
Abstract
The objective of this study was to evaluate the digestive tract recovery and metabolism of feeding either bovine colostrum (BC), transition milk (TM), or milk replacer (MR) after an episode of feed restriction and fasting (FRF) in dairy calves. Thirty-five Holstein male calves (22 ± 4.8 d old) were involved in a 50-d study. After 3 d of feeding 2 L of rehydration solution twice daily and 19 h of fasting (d 1 of study), calves were randomly assigned to one of the 5 feeding treatments (n = 7): calves were offered either pooled BC during 4 (C4) or 10 (C10) days, pooled TM during 4 (TM4) or 10 (TM10) days, or MR for 10 d (CTRL) at the rate of 720 g/d DM content. Then, all calves were fed the same feeding program, gradually decreasing MR from 3 L twice daily to 2 L once daily at 12.5% DM until weaning (d 42), and concentrate feed, water, and straw were offered ad libitum until d 50. Citrulline, Cr-EDTA, β-hydroxybutyrate (BHB), and nonesterified fatty acids (NEFA) in serum and complete blood count (CBC) were determined on d -3, 1, 2, 5, and 11 relative to FRF, except BHB and NEFA at d -3. Volatile fatty acids (VFA), lactoferrin (LTF), IgA, and microbiota (Firmicutes to Bacteroidetes ratio and Fecalis prausnitzii) were analyzed in feces on d 5 and 11 before the morning feeding. Health scores were recorded daily from d -3 to d 14 as well as d 23 and 30. Feed concentrate, MR, and straw intake were recorded daily, and body weight on d -3, 1, 2, 5, and 11 and weekly afterward. Calf performance, intake, serum Cr-EDTA, CBC, fecal LTF concentrations and microbiota parameters were similar among treatments throughout the study. Serum NEFA concentrations were greater in TM4, TM10 and C10 calves compared with the CTRL ones from d 2 to 11, and after the FRF, serum concentrations of BHB were lower in CTRL calves than in the other treatments, and on d 11, serum BHB concentrations in the long treatments (C10 and TM10) remained greater than those in the shorter ones (C4 and TM4) and CTRL. Serum citrulline concentrations were similar on d -3 and 1 in all treatments, but they were greater in C4, C10, TM4, and TM10 on d 2 and 5, and on d 11 they were only greater in C10 and TM10 than in CTRL calves. Fecal IgA concentrations tended to be greater in C10 than in CTRL, TM4, and TM10 calves, and in C4 and TM10 than in CTRL animals. Fecal propionate proportion was lesser in C10 than in CTRL, TM4, and TM10 calves, while butyrate was greater in C4 and C10 than in TM4 and CTRL calves. The proportion of non-normal fecal scores of C10 fed calves was greater than TM4 and TM10 calves. Results showed that TM and BC may help to recover intestinal functionality, provide gut immune protection, and increase liver fatty acid oxidation in calves after a FRF episode.
Collapse
Affiliation(s)
- M Tortadès
- Department of Ruminant Production, IRTA (Institut de Recerca i Tecnologia Agroalimentàries), 08140 Caldes de Montbui, Barcelona, Spain
| | - S Marti
- Department of Ruminant Production, IRTA (Institut de Recerca i Tecnologia Agroalimentàries), 08140 Caldes de Montbui, Barcelona, Spain
| | - M Devant
- Department of Ruminant Production, IRTA (Institut de Recerca i Tecnologia Agroalimentàries), 08140 Caldes de Montbui, Barcelona, Spain
| | - M Vidal
- Department of Ruminant Production, IRTA (Institut de Recerca i Tecnologia Agroalimentàries), 08140 Caldes de Montbui, Barcelona, Spain
| | - F Fàbregas
- Department of Ruminant Production, IRTA (Institut de Recerca i Tecnologia Agroalimentàries), 08140 Caldes de Montbui, Barcelona, Spain
| | - M Terré
- Department of Ruminant Production, IRTA (Institut de Recerca i Tecnologia Agroalimentàries), 08140 Caldes de Montbui, Barcelona, Spain.
| |
Collapse
|
87
|
Yin P, Du T, Yi S, Zhang C, Yu L, Tian F, Chen W, Zhai Q. Response differences of gut microbiota in oligofructose and inulin are determined by the initial gut Bacteroides/Bifidobacterium ratios. Food Res Int 2023; 174:113598. [PMID: 37986462 DOI: 10.1016/j.foodres.2023.113598] [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: 07/30/2023] [Revised: 10/07/2023] [Accepted: 10/13/2023] [Indexed: 11/22/2023]
Abstract
Prebiotics are known to modulate the gut microbiota, but there is host variability, mainly due to differences in carbohydrate-utilisation by gut microbiota. Bifidobacterium and Bacteroides are powerful carbohydrate-utilising bacteria, and the ratio of both is closely related to the utilisation of prebiotics. However, the differential impact of prebiotics on the composition and function of the gut microbiota and its metabolites in participants with different Bacteroides/Bifidobacterium (Ba/Bi) ratios have not been studied. Here, we conducted a 4-week randomised double-blind, parallel four-arm trial using two prebiotics (oligofructose and inulin) in two populations with high Ba/Bi (H) and low Ba/Bi (L). The response to prebiotics in both populations was influenced by the baseline microbiota background specificity. Notably, at an overall level, FOS was slightly better than inulin in modulating the gut microbiota. Difference in gut microbiota regulation by FOS across microbiota contexts were significant between the two groups. Butyric acid-producing bacteria were significantly more abundant in H and further elevated butyric acid and related metabolite levels, with H more likely to benefit from the FOS intervention. The two groups showed only metabolic differences in their response to inulin, with L showing a significant increase in propionic acid and being enriched in glycolysis functions, whereas H was enriched in amino acids and aminoglycolysis functions. Overall, these results provide a basis for selecting appropriate prebiotics for participants with different gut backgrounds.
Collapse
Affiliation(s)
- Pingping Yin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ting Du
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Shanrong Yi
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Chengcheng Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
88
|
Guo Z, Yiu N, Hu Z, Zhou W, Long X, Yang M, Liao J, Zhang G, Lu Q, Zhao M. Alterations of fecal microbiome and metabolome in pemphigus patients. J Autoimmun 2023; 141:103108. [PMID: 37714737 DOI: 10.1016/j.jaut.2023.103108] [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: 07/12/2023] [Accepted: 08/24/2023] [Indexed: 09/17/2023]
Abstract
The role of gut microbiome and metabolic substances in the development of autoimmune diseases has gradually been revealed. However, the relevant gut features in pemphigus have not been well clarified. We collected stool samples from pemphigus patients and healthy controls (HCs). Metagenomic sequencing and liquid chromatography-mass spectrometry (LC/MS) metabolome sequencing were performed to analyze the compositional and metabolic alternations of the gut microbiome in pemphigus patients and HCs. We observed the reduced richness and diversity and greater heterogeneity in pemphigus patients, which was characterized by a significant decrease in Firmicutes and an increase in Proteobacteria. At the species level, Intestinal pathogenic bacteria such as Escherichia coli and Bacteroides fragilis were significantly enriched, while anti-inflammatory bacteria and butyric acid-producing bacteria were significantly reduced, which were related to clinical indicators (Dsg1/3 and PDAI). 4 species were selected by the machine learning algorithm to better distinguish pemphigus patients from healthy people. Metabolomic analysis showed that the composition of pemphigus patients was different from that of HCs. PE (18:3 (6Z,9Z, 12Z)/14:1 (9Z)) was the main metabolic substance in pemphigus and involved in a variety of metabolic pathways. While Retinol, flavonoid compounds and various amino acids decreased significantly compared with HCs. Furthermore, we found that differences in the levels of these metabolites correlated with changes in the abundance of specific species. Our study provides a comprehensive picture of gut microbiota and metabolites in pemphigus patients and suggests a potential mechanism of the aberrant gut microbiota and metabolites in the pathogenesis of pemphigus.
Collapse
Affiliation(s)
- Ziyu Guo
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Nam Yiu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Zhi Hu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Wenyu Zhou
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Xuan Long
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Miao Yang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Jieyue Liao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Guiying Zhang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China.
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
| |
Collapse
|
89
|
Song WS, Jo SH, Lee JS, Kwon JE, Park JH, Kim YR, Baek JH, Kim MG, Kwon SY, Kim YG. Multiomics analysis reveals the biological effects of live Roseburia intestinalis as a high-butyrate-producing bacterium in human intestinal epithelial cells. Biotechnol J 2023; 18:e2300180. [PMID: 37596881 DOI: 10.1002/biot.202300180] [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: 04/24/2023] [Revised: 07/29/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
Butyrate-producing bacteria play a key role in human health, and recent studies have triggered interest in their development as next-generation probiotics. However, there remains limited knowledge not only on the identification of high-butyrate-producing bacteria in the human gut but also in the metabolic capacities for prebiotic carbohydrates and their interaction with the host. Herein, it was discovered that Roseburia intestinalis produces higher levels of butyrate and digests a wider variety of prebiotic polysaccharide structures compared with other human major butyrate-producing bacteria (Eubacterium rectale, Faecalibacterium prausnitzii, and Roseburia hominis). Moreover, R. intestinalis extracts upregulated the mRNA expression of tight junction proteins (TJP1, OCLN, and CLDN3) in human intestinal epithelial cells more than other butyrate-producing bacteria. R. intestinalis was cultured with human intestinal epithelial cells in the mimetic intestinal host-microbe interaction coculture system to explore the health-promoting effects using multiomics approaches. Consequently, it was discovered that live R. intestinalis only enhances purine metabolism and the oxidative pathway, increasing adenosine triphosphate levels in human intestinal epithelial cells, but that heat-killed bacteria had no effect. Therefore, this study proposes that R. intestinalis has potentially high value as a next-generation probiotic to promote host intestinal health.
Collapse
Affiliation(s)
- Won-Suk Song
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Sung-Hyun Jo
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Jae-Seung Lee
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Ji-Eun Kwon
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Ji-Hyeon Park
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Ye-Rim Kim
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Ji-Hyun Baek
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Min-Gyu Kim
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Seo-Young Kwon
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Yun-Gon Kim
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| |
Collapse
|
90
|
Li J, Ma J, Wang W, Du H, Tang S, Li Y, Zhu W, Zhang R, Wan J. Alterations of ileal mucosa-associated microbiota in hypercholesterolemia patients. Heliyon 2023; 9:e22116. [PMID: 38076161 PMCID: PMC10709208 DOI: 10.1016/j.heliyon.2023.e22116] [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: 06/17/2023] [Revised: 09/11/2023] [Accepted: 11/05/2023] [Indexed: 01/03/2025] Open
Abstract
Many metabolic diseases have been demonstrated to be associated with changes in the microbiome. However, no studies have yet been conducted to examine the characteristics of the mucosal microbiota of patients with hypercholesterolemia. We aimed to examine mucosa-associated microbiota in subjects with hypercholesterolemia. We conducted a case-control study, in which ileal mucosal samples were collected from 13 hypercholesterolemia patients and 13 controls for 16S rRNA gene sequencing. There were differences in the composition of ileal mucosal microbiota based on beta diversity between the hypercholesterolemia and control groups (P < 0.05). Compared with the control group, the phylum Bacteroidetes and the genera Bacteroides, Butyricicoccus, Parasutterella, Candidatus_Soleaferrea, and norank_f__norank_o__Izemoplasmatales were less abundant in the hypercholesterolemia group (P < 0.05), while the genus Anaerovibrio was enriched in the hypercholesterolemia group (P < 0.05). The relative abundance of Bacteroides was negatively correlated with total cholesterol and low-density lipoprotein cholesterol (P < 0.01). The relative abundance of Coprococcus was negatively correlated with triglycerides and body mass index (all P < 0.05). PICRUSt functional prediction analysis showed that pathways related to Glycerophospholipid metabolism, ABC transporters, Phosphotransferase system, and Biofilm formation - Escherichia coli, and infectious diseases of pathogenic Escherichia coli were enriched in the hypercholesterolemia group. This work suggests a potential role of ileal mucosal microbiota in the development of hypercholesterolemia.
Collapse
Affiliation(s)
- Jia Li
- Department of Gastroenterology, The 983rd Hospital of Joint Logistic Support Force of PLA, No. 60, Huangwei Road, Hebei District, Tianjin 300142, China
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Jinxia Ma
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Weihua Wang
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Haitao Du
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Shuai Tang
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Yi Li
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Wenya Zhu
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Ru Zhang
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Jun Wan
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| |
Collapse
|
91
|
Ye Q, Sun S, Deng J, Chen X, Zhang J, Lin S, Du H, Gao J, Zou X, Lin X, Cai Y, Lu Z. Using 16S rDNA and metagenomic sequencing technology to analyze the fecal microbiome of children with avoidant/restrictive food intake disorder. Sci Rep 2023; 13:20253. [PMID: 37985845 PMCID: PMC10661725 DOI: 10.1038/s41598-023-47760-y] [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: 08/16/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023] Open
Abstract
To investigate the gut microbiota distribution and its functions in children with avoidant/restrictive food intake disorder (ARFID). A total of 135 children were enrolled in the study, including 102 children with ARFID and 33 healthy children. Fecal samples were analyzed to explore differences in gut microbiota composition and diversity and functional differences between the ARFID and healthy control (HC) groups via 16S rDNA and metagenomic sequencing. The gut microbiota composition and diversity in children with ARFID were different from those in heathy children, but there is no difference in the composition and diversity of gut microbiota between children at the age of 3-6 and 7-12 with ARFID. At the phylum level, the most abundant microbes in the two groups identified by 16S rDNA and metagenomic sequencing were the same. At the genus level, the abundance of Bacteroides was higher in the ARFID group (P > 0.05); however, different from the result of 16SrDNA sequencing, metagenomic sequencing showed that the abundance of Bacteroides in the ARFID group was significantly higher than that in the HC group (P = 0.041). At the species level, Escherichia coli, Streptococcus thermophilus and Lachnospira eligens were the most abundant taxa in the ARFID group, and Prevotella copri, Bifidobacterium pseudocatenulatum, and Ruminococcus gnavus were the top three microbial taxa in the HC group; there were no statistically significant differences between the abundance of these microbial taxa in the two groups. LefSe analysis indicated a greater abundance of the order Enterobacterales and its corresponding family Enterobacteriaceae, the family Bacteroidaceae and corresponding genus Bacteroides, the species Bacteroides vulgatus in ARFID group, while the abundance of the phylum Actinobacteriota and its corresponding class Actinobacteria , the order Bifidobacteriales and corresponding family Bifidobacteriaceae, the genus Bifidobacterium were enriched in the HC group. There were no statistically significant differences in the Chao1, Shannon and Simpson indices between the Y1 and Y2 groups (P = 0.1, P = 0.06, P = 0.06). At the phylum level, Bacillota, Bacteroidota, Proteobacteria and Actinobacteriota were the most abundant taxa in both groups, but there were no statistically significant differences among the abundance of these bacteria (P = 0.958, P = 0.456, P = 0.473, P = 0.065). At the genus level, Faecalibacterium was more abundant in the Y2 group than in the Y1 group, and the difference was statistically significant (P = 0.037). The KEGG annotation results showed no significant difference in gut microbiota function between children with ARFID and healthy children; however, GT26 was significantly enriched in children with ARFID based on the CAZy database. The most abundant antibiotic resistance genes in the ARFID group were the vanT, tetQ, adeF, ermF genes, and the abundance of macrolide resistance genes in the ARFID group was significantly higher than that in the HC group (P = 0.041). Compared with healthy children, children with ARFID have a different distribution of the gut microbiota and functional genes. This indicates that the gut microbiome might play an important role in the pathogenesis of ARFID.Clinical trial registration: ChiCTR2300074759.
Collapse
Affiliation(s)
- Qina Ye
- Department of Traditional Chinese Medicine, Guangzhou Women and Children Medical Center, No. 9 Jinsui Road, Guangzhou, 510623, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Shaodan Sun
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jian Deng
- Department of Traditional Chinese Medicine, Guangzhou Women and Children Medical Center, No. 9 Jinsui Road, Guangzhou, 510623, China
| | - Xiaogang Chen
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jing Zhang
- Department of Traditional Chinese Medicine, Guangzhou Women and Children Medical Center, No. 9 Jinsui Road, Guangzhou, 510623, China
| | - Suihua Lin
- Department of Traditional Chinese Medicine, Guangzhou Women and Children Medical Center, No. 9 Jinsui Road, Guangzhou, 510623, China
| | - Hongxuan Du
- Department of Traditional Chinese Medicine, Guangzhou Women and Children Medical Center, No. 9 Jinsui Road, Guangzhou, 510623, China
| | - Jinxiong Gao
- Department of Traditional Chinese Medicine, Guangzhou Women and Children Medical Center, No. 9 Jinsui Road, Guangzhou, 510623, China
| | - Xiaoyin Zou
- Department of Traditional Chinese Medicine, Guangzhou Women and Children Medical Center, No. 9 Jinsui Road, Guangzhou, 510623, China
| | - Xiaoling Lin
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yawen Cai
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhuoming Lu
- Department of Traditional Chinese Medicine, Guangzhou Women and Children Medical Center, No. 9 Jinsui Road, Guangzhou, 510623, China.
| |
Collapse
|
92
|
Hou J, Lian L, Lu L, Gu T, Zeng T, Chen L, Xu W, Li G, Wu H, Tian Y. Effects of Dietary Bacillus coagulans and Tributyrin on Growth Performance, Serum Antioxidants, Intestinal Morphology, and Cecal Microbiota of Growing Yellow-Feathered Broilers. Animals (Basel) 2023; 13:3534. [PMID: 38003151 PMCID: PMC10668748 DOI: 10.3390/ani13223534] [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: 10/08/2023] [Revised: 10/30/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
This study investigated the impact of Bacillus coagulans (BC) and tributyrin (TB) supplementation on the growth performance, serum antioxidant capacity, intestinal morphology, and cecal microbiota of yellow-feathered broilers. Using a 2 × 2 factorial design, 480 broilers were randomly assigned to four experimental diets, comprising two levels of BC (0 and 1 g/kg) and two levels of TB (0 and 1 g/kg), over a 36-day period. A significant interaction was observed between BC and TB, impacting the average daily feed intake (ADFI) of broilers aged between 26 and 40 days (p < 0.01). BC and TB also displayed a significant interaction in relation to serum malondialdehyde levels and total antioxidant capacity (p < 0.05). Additionally, there was a significant interaction between BC and TB concerning the duodenal villus-to-crypt ratio, crypt depth, and jejunal villus-to-crypt ratio (p < 0.05). The addition of BC and TB significantly enhanced the richness and diversity of cecal microbiota, with a notable interactive effect observed for the abundance of Faecalibacterium, Ruminococcus_torques_group, and Phascolarctobacterium. In conclusion, supplementation with BC and TB can effectively improve the growth performance, serum antioxidant capacity, intestinal morphology, and cecal microbiota composition of yellow-feathered broilers, indicating the presence of an interactive effect.
Collapse
Affiliation(s)
- Jinwang Hou
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| | - Lina Lian
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| | - Lizhi Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| | - Tiantian Gu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| | - Tao Zeng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| | - Li Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| | - Wenwu Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| | - Guoqin Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| | - Hongzhi Wu
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Yong Tian
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| |
Collapse
|
93
|
Feng Y, Xu D. Short-chain fatty acids are potential goalkeepers of atherosclerosis. Front Pharmacol 2023; 14:1271001. [PMID: 38027009 PMCID: PMC10679725 DOI: 10.3389/fphar.2023.1271001] [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: 08/02/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Short-chain fatty acids (SCFAs) are metabolites produced by gut bacteria and play a crucial role in various inflammatory diseases. Increasing evidence suggests that SCFAs can improve the occurrence and progression of atherosclerosis. However, the molecular mechanisms through which SCFAs regulate the development of atherosclerosis have not been fully elucidated. This review provides an overview of the research progress on SCFAs regarding their impact on the risk factors and pathogenesis associated with atherosclerosis, with a specific focus on their interactions with the endothelium and immune cells. These interactions encompass the inflammation and oxidative stress of endothelial cells, the migration of monocytes/macrophages, the lipid metabolism of macrophages, the proliferation and migration of smooth muscle cells, and the proliferation and differentiation of Treg cells. Nevertheless, the current body of research is insufficient to comprehensively understand the full spectrum of SCFAs' mechanisms of action. Therefore, further in-depth investigations are imperative to establish a solid theoretical foundation for the development of clinical therapeutics in this context.
Collapse
Affiliation(s)
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
94
|
Fan L, Xia Y, Wang Y, Han D, Liu Y, Li J, Fu J, Wang L, Gan Z, Liu B, Fu J, Zhu C, Wu Z, Zhao J, Han H, Wu H, He Y, Tang Y, Zhang Q, Wang Y, Zhang F, Zong X, Yin J, Zhou X, Yang X, Wang J, Yin Y, Ren W. Gut microbiota bridges dietary nutrients and host immunity. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2466-2514. [PMID: 37286860 PMCID: PMC10247344 DOI: 10.1007/s11427-023-2346-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 04/05/2023] [Indexed: 06/09/2023]
Abstract
Dietary nutrients and the gut microbiota are increasingly recognized to cross-regulate and entrain each other, and thus affect host health and immune-mediated diseases. Here, we systematically review the current understanding linking dietary nutrients to gut microbiota-host immune interactions, emphasizing how this axis might influence host immunity in health and diseases. Of relevance, we highlight that the implications of gut microbiota-targeted dietary intervention could be harnessed in orchestrating a spectrum of immune-associated diseases.
Collapse
Affiliation(s)
- Lijuan Fan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yaoyao Xia
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Youxia Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Jiahuan Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jie Fu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Leli Wang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Zhending Gan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Bingnan Liu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jian Fu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Congrui Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Zhenhua Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jinbiao Zhao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Hui Han
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Hao Wu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yiwen He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Yulong Tang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Qingzhuo Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yibin Wang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Fan Zhang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Xin Zong
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
| | - Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China.
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
| | - Wenkai Ren
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
95
|
Sun Y, Zhang S, Nie Q, He H, Tan H, Geng F, Ji H, Hu J, Nie S. Gut firmicutes: Relationship with dietary fiber and role in host homeostasis. Crit Rev Food Sci Nutr 2023; 63:12073-12088. [PMID: 35822206 DOI: 10.1080/10408398.2022.2098249] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Firmicutes and Bacteroidetes are the predominant bacterial phyla colonizing the healthy human gut. Accumulating evidence suggests that dietary fiber plays a crucial role in host health, yet most studies have focused on how the dietary fiber affects health through gut Bacteroides. More recently, gut Firmicutes have been found to possess many genes responsible for fermenting dietary fiber, and could also interact with the intestinal mucosa and thereby contribute to homeostasis. Consequently, the relationship between dietary fiber and Firmicutes is of interest, as well as the role of Firmicutes in host health. In this review, we summarize the current knowledge regarding the molecular mechanism of dietary fiber degradation by gut Firmicutes and explain the communication pathway of the dietary fiber-Firmicutes-host axis, and the beneficial effects of dietary fiber-induced Firmicutes and their metabolites on health. A better understanding of the dialogue sustained by the dietary fiber-Firmicutes axis and the host could provide new insights into probiotic therapy and novel dietary interventions aimed at increasing the abundance of Firmicutes (such as Faecalibacterium, Lactobacillus, and Roseburia) to promote health.
Collapse
Affiliation(s)
- Yonggan Sun
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Shanshan Zhang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Qixing Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Huijun He
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Huizi Tan
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Fang Geng
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Haihua Ji
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Jielun Hu
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| |
Collapse
|
96
|
Rodriquez-Saavedra M, Tamargo A, Molinero N, Relaño de la Guía E, Jiménez-Arroyo C, Bartolomé B, González de Llano D, Victoria Moreno-Arribas M. Simulated gastrointestinal digestion of beer using the simgi® model. Investigation of colonic phenolic metabolism and impact on human gut microbiota. Food Res Int 2023; 173:113228. [PMID: 37803545 DOI: 10.1016/j.foodres.2023.113228] [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: 03/07/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 10/08/2023]
Abstract
Beer is a source of bioactive compounds, mainly polyphenols, which can reach the large intestine and interact with colonic microbiota. However, the effects of beer consumption in the gastrointestinal function have scarcely been studied. This paper reports, for the first time, the in vitro digestion of beer and its impact on intestinal microbiota metabolism. Three commercial beers of different styles were subjected to gastrointestinal digestion using the simgi® model, and the digested fluids were further fermented in triplicate with faecal microbiota from a healthy volunteer. The effect of digested beer on human gut microbiota was evaluated in terms of microbial metabolism (short-chain fatty acids (SCFAs) and ammonium ion), microbial diversity and bacterial populations (plate counting and 16S rRNA gene sequencing). Monitoring beer polyphenols through the different digestion phases showed their extensive metabolism, mainly at the colonic stage. In addition, a higher abundance of taxa related to gut health, especially Bacteroides, Bifidobacterium, Mitsuokella and Succinilasticum at the genus level, and the Ruminococcaceae and Prevotellaceae families were found in the presence of beers. Regarding microbial metabolism, beer feeding significantly increased microbial SCFA production (mainly butyric acid) and decreased ammonium content. Overall, these results evidence the positive actions of moderate beer consumption on the metabolic activity of colonic microbiota, suggesting that the raw materials and brewing methods used may affect the beer gut effects.
Collapse
Affiliation(s)
| | - Alba Tamargo
- Institute of Food Science Research (CIAL), CSIC-UAM, c/ Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Natalia Molinero
- Institute of Food Science Research (CIAL), CSIC-UAM, c/ Nicolás Cabrera 9, 28049 Madrid, Spain
| | | | - Cristina Jiménez-Arroyo
- Institute of Food Science Research (CIAL), CSIC-UAM, c/ Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Begoña Bartolomé
- Institute of Food Science Research (CIAL), CSIC-UAM, c/ Nicolás Cabrera 9, 28049 Madrid, Spain
| | | | | |
Collapse
|
97
|
Xu J, Wang R, Liu W, Yin Z, Wu J, Yu X, Wang W, Zhang H, Li Z, Gao M, Zhu L, Zhan X. The specificity of ten non-digestible carbohydrates to enhance butyrate-producing bacteria and butyrate production in vitro fermentation. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2023.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
98
|
Klostermann CE, Endika MF, Ten Cate E, Buwalda PL, de Vos P, Bitter JH, Zoetendal EG, Schols HA. Type of intrinsic resistant starch type 3 determines in vitro fermentation by pooled adult faecal inoculum. Carbohydr Polym 2023; 319:121187. [PMID: 37567720 DOI: 10.1016/j.carbpol.2023.121187] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 08/13/2023]
Abstract
Resistant starch (RS) results in relatively high health-beneficial butyrate levels upon fermentation by gut microbiota. We studied how physico-chemical characteristics of RS-3 influenced butyrate production during fermentation. Six highly resistant RS-3 substrates (intrinsic RS-3, 80-95 % RS) differing in chain length (DPn 16-76), Mw distribution (PI) and crystal type (A/B) were fermented in vitro by pooled adult faecal inoculum. All intrinsic RS-3 substrates were fermented to relatively high butyrate levels (acetate/butyrate ≤ 2.5), and especially fermentation of A-type RS-3 prepared from polydisperse α-1,4 glucans resulted in the highest relative butyrate amount produced (acetate/butyrate: 1). Analysis of the microbiota composition after fermentation revealed that intrinsic RS-3 stimulated primarily Lachnospiraceae, Bifidobacterium and Ruminococcus, but the relative abundances of these taxa differed slightly depending on the RS-3 physico-chemical characteristics. Especially intrinsic RS-3 of narrow disperse Mw distribution stimulated relatively more Ruminococcus. Selected RS fractions (polydisperse Mw distribution) obtained after pre-digestion were fermented to acetate and butyrate (ratio ≤ 1.8) and stimulated Lachnospiraceae and Bifidobacterium. This study indicates that especially the α-1,4 glucan Mw distribution dependent microstructure of RS-3 influences butyrate production and microbiota composition during RS-3 fermentation.
Collapse
Affiliation(s)
- C E Klostermann
- Biobased Chemistry and Technology, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, the Netherlands; Laboratory of Food Chemistry, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, the Netherlands
| | - M F Endika
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - E Ten Cate
- Biobased Chemistry and Technology, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, the Netherlands; Laboratory of Food Chemistry, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, the Netherlands
| | - P L Buwalda
- Biobased Chemistry and Technology, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, the Netherlands; Coöperatie Koninklijke AVEBE u.a., P.O. Box 15, 9640 AA Veendam, the Netherlands
| | - P de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Centre Groningen, Groningen, Hanzeplein 1, 9700 RB Groningen, the Netherlands
| | - J H Bitter
- Biobased Chemistry and Technology, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, the Netherlands
| | - E G Zoetendal
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - H A Schols
- Laboratory of Food Chemistry, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, the Netherlands.
| |
Collapse
|
99
|
Yao C, Gou X, Tian C, Zhou L, Hao R, Wan L, Wang Z, Li M, Tong X. Key regulators of intestinal stem cells: diet, microbiota, and microbial metabolites. J Genet Genomics 2023; 50:735-746. [PMID: 36566949 DOI: 10.1016/j.jgg.2022.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Interactions between diet and the intestinal microbiome play an important role in human health and disease development. It is well known that such interactions, whether direct or indirect, trigger a series of metabolic reactions in the body. Evidence suggests that intestinal stem cells (ISCs), which are phenotypic precursors of various intestinal epithelial cells, play a significant role in the regulation of intestinal barrier function and homeostasis. The advent and evolution of intestinal organoid culture techniques have presented a key opportunity to study the association between the intestinal microenvironment and ISCs. As a result, the effects exerted by dietary factors, intestinal microbiomes, and their metabolites on the metabolic regulation of ISCs and the potential mechanisms underlying such effects are being gradually revealed. This review summarises the effects of different dietary patterns on the behaviour and functioning of ISCs and focuses on the crosstalk between intestinal microbiota, related metabolites, and ISCs, with the aim of fully understanding the relationship between these three factors and providing further insights into the complex mechanisms associated with ISCs in the human body. Gaining an understanding of these mechanisms may lead to the development of novel dietary interventions or drugs conducive to intestinal health.
Collapse
Affiliation(s)
- Chensi Yao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xiaowen Gou
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Chuanxi Tian
- Graduate College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Lijuan Zhou
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Rui Hao
- Graduate College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Li Wan
- Graduate College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zeyu Wang
- Department of Scientific Research, Changchun University of Chinese Medicine, Changchun, Jilin 130017, China.
| | - Min Li
- Molecular Biology Laboratory, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Xiaolin Tong
- Institute of Metabolic Diseases, Guang'anmen Hospital of China, Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
100
|
Li L, Yan S, Liu S, Wang P, Li W, Yi Y, Qin S. In-depth insight into correlations between gut microbiota and dietary fiber elucidates a dietary causal relationship with host health. Food Res Int 2023; 172:113133. [PMID: 37689844 DOI: 10.1016/j.foodres.2023.113133] [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: 04/03/2023] [Revised: 06/09/2023] [Accepted: 06/10/2023] [Indexed: 09/11/2023]
Abstract
Dietary fiber exerts a wide range of biological benefits on host health, which not only provides a powerful source of nutrition for gut microbiota but also supplies key microbial metabolites that directly affect host health. This review mainly focuses on the decomposition and metabolism of dietary fiber and the essential genera Bacteroides and Bifidobacterium in dietary fiber fermentation. Dietary fiber plays an essential role in host health by impacting outcomes related to obesity, enteritis, immune health, cancer and neurodegenerative diseases. Additionally, the gut microbiota-independent pathway of dietary fiber affecting host health is also discussed. Personalized dietary fiber intake combined with microbiome, genetics, epigenetics, lifestyle and other factors has been highlighted for development in the future. A higher level of evidence is needed to demonstrate which microbial phenotype benefits from which kind of dietary fiber. In-depth insights into the correlation between gut microbiota and dietary fiber provide strong theoretical support for the precise application of dietary fiber, which elucidates a dietary causal relationship with host health.
Collapse
Affiliation(s)
- Lili Li
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| | - Shuling Yan
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuangjiang Liu
- Shandong University, Qingdao 266237, China; Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Ping Wang
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Wenjun Li
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| | - Yuetao Yi
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| | - Song Qin
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| |
Collapse
|