1
|
Kumar S, Mukherjee R, Gaur P, Leal É, Lyu X, Ahmad S, Puri P, Chang CM, Raj VS, Pandey RP. Unveiling roles of beneficial gut bacteria and optimal diets for health. Front Microbiol 2025; 16:1527755. [PMID: 40041870 PMCID: PMC11877911 DOI: 10.3389/fmicb.2025.1527755] [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: 11/18/2024] [Accepted: 02/03/2025] [Indexed: 04/02/2025] Open
Abstract
The gut microbiome plays a pivotal role in human health, influencing digestion, immunity, and disease prevention. Beneficial gut bacteria such as Akkermansia muciniphila, Adlercreutzia equolifaciens, and Christensenella minuta contribute to metabolic regulation and immune support through bioactive metabolites like short-chain fatty acids (SCFAs). Dietary patterns rich in prebiotics, fermented foods, and plant-based bioactive compounds, including polyphenols and flavonoids, promote microbiome diversity and stability. However, challenges such as individual variability, bioavailability, dietary adherence, and the dynamic nature of the gut microbiota remain significant. This review synthesizes current insights into gut bacteria's role in health, emphasizing the mechanisms by which dietary interventions modulate microbiota. Additionally, it highlights advancements in microbiome-targeted therapies and the transformative potential of personalized nutrition, leveraging microbiota profiling and artificial intelligence (AI) to develop tailored dietary strategies for optimizing gut health and mitigating chronic inflammatory disorders. Addressing these challenges requires a multidisciplinary approach that integrates scientific innovation, ethical frameworks, and practical implementation strategies.
Collapse
Affiliation(s)
- Suresh Kumar
- National Institute of Biologicals, Ministry of Health & Family Welfare, Govt. of India, Noida, India
| | - Riya Mukherjee
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, Taiwan
| | - Pratibha Gaur
- Centre for Drug Design Discovery and Development (C4D), SRM University Delhi-NCR, Sonepat, India
- Department of Biotechnology and Microbiology, SRM University Delhi-NCR, Sonepat, India
| | - Élcio Leal
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, Brazil
| | - Xiaoming Lyu
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Saheem Ahmad
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Ha'il, Saudi Arabia
| | - Paridhi Puri
- University Centre for Research and Development, Chandigarh University, Mohali, India
| | - Chung-Ming Chang
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, Taiwan
- Master & Ph.D Program in Biotechnology Industry, Chang Gung University, Taoyuan, Taiwan
| | - V. Samuel Raj
- Department of Biotechnology and Microbiology, SRM University Delhi-NCR, Sonepat, India
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, Brazil
| | - Ramendra Pati Pandey
- Department of Biotechnology and Microbiology, SRM University Delhi-NCR, Sonepat, India
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, Brazil
| |
Collapse
|
2
|
Gayathri K, Nalini R, Sriram S, Jayaraj I, Parvathi VD, Velraja S. Microbiological Profile of Phytoestrogen Rich Supplement and Its Impact on Gut-microbiome Composition in Drosophila melanogaster. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2024; 18:2005-2013. [DOI: 10.22207/jpam.18.3.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
Phytoestrogen-rich diet alters the composition of gut microbiota by enhancing the growth of beneficial bacteria and decreasing the microbial load of pathogenic organisms. Drosophila is an invertebrate model system used for research studies, as it shares 70% genetic homology with humans. The present study aimed to analyse microbiological profile of phytoestrogen rich supplement and its impact on gut-microbiome composition in Drosophila melanogaster. The phytoestrogen rich supplement was mixed with formula 424 plain and flies were exposed to it. Gut of flies was dissected and cell suspension was prepared. Bacterial colonies were developed by streaking method. Gram staining was performed to differentiate the bacterial cells and further gut microbiome composition (Acetobacteraceae and Lactobacillales taxa) was analysed by 16S rRNA sequencing. The microbiological analysis was carried out to ascertain the microbial load of the developed product for consumption. The total bacterial count and coliform counts of the phytoestrogen rich supplement were <10 CFU/g. Also, the developed supplement exhibited minimal yeast and mold growth (<1 CFU/g). Gram staining showed gram positive (Bacilli and cocci). 16S rRNA sequencing showed significance with mild variation in similarity. It confirmed the presence of Bacillus paramycoids. The developed supplement has showed improved gut microbiome composition in the Drosophila. In future, studies can be extended to humans to analyse the efficacy of the supplement in the gut microbiome composition.
Collapse
|
3
|
Vanslette AM, Toft PB, Lund ML, Moritz T, Arora T. Serotonin receptor 4 agonism prevents high fat diet induced reduction in GLP-1 in mice. Eur J Pharmacol 2023; 960:176181. [PMID: 37926275 DOI: 10.1016/j.ejphar.2023.176181] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 11/07/2023]
Abstract
Hormone-producing enteroendocrine cells (EECs) are present throughout the gastrointestinal tract and respond to various nutrient and gut microbiota produced metabolites stimuli. Two important EEC subtypes, Glucagon like peptide-1 (GLP-1) producing L-cells and serotonin (5-HT) producing enterochromaffin (EC) cells interact via paracrine signaling and exhibit bidirectional regulation of expression and secretion of produced hormones. Accordingly, in vitro studies suggest potential to modulate 5-HT secretion by GLP-1 receptor agonism, and L-cell differentiation via serotonin receptor 4 agonism. However, the importance of this cellular signaling on host metabolism is poorly understood. In this study, we found that two weeks of high fat diet (HFD) feeding reduced RNA expression of gut hormones, including proglucagon (Gcg) gene encoding GLP-1 and Tryptophan hydroxylase1 (Tph1) gene encoding rate limiting enzyme in 5-HT synthesis, specifically in the colon and reduced plasma GLP-1 levels. Levels of propionate and butyrate were also reduced following HFD. However, supplementation of sodium propionate did not improve HFD induced reduction in GLP-1. In contrast, chemical induction of serotonin receptor 4 promoted GLP-1 levels, colonic Gcg RNA expression accompanied by improvement in glucose tolerance in HFD-fed mouse. Thus, this study suggests a novel mechanism to improve glucose tolerance via serotonin receptor 4 stimulation in the HFD induced obese mouse model.
Collapse
Affiliation(s)
- Amanda Marie Vanslette
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Pernille Baumann Toft
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Mari Lilith Lund
- Human Health Research, Scientific Affairs, Chr. Hansen A/S, Bøge Alle 10-12, 2970, Hørsholm, Denmark
| | - Thomas Moritz
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Tulika Arora
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| |
Collapse
|
4
|
Wang D, Ma B, Liao Z, Li W, Zhang T, Lei C, Wang H. Flaxseed Supplementation in Chicken Feed Accelerates Salmonella enterica subsp. enterica Serovar Enteritidis Clearance, Modulates Cecum Microbiota, and Influences Ovarian Gene Expression in Laying Hens. Biomolecules 2023; 13:1353. [PMID: 37759753 PMCID: PMC10526464 DOI: 10.3390/biom13091353] [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: 07/04/2023] [Revised: 08/18/2023] [Accepted: 08/20/2023] [Indexed: 09/29/2023] Open
Abstract
Salmonella is a foodborne pathogen that poses a serious threat to both human and animal health and food safety. Flaxseed is rich in unsaturated fatty acids; has anti-metabolic syndrome, anti-inflammatory, and neuroprotective properties; and may be a potential source of feed additives. To investigate the impact of flaxseed on Salmonella-infected laying hens, we administered Salmonella enterica subsp. enterica serovar Enteritidis (S. Enteritidis) after adding flaxseed to the feed of laying hens (15% [750 mg/kg]). S. Enteritidis colonization was reduced and its clearance was accelerated from the laying hens. Furthermore, flaxseed supplementation mitigated the damage to the ileum caused by S. Enteritidis. We analyzed alterations in intestinal flora through 16S rRNA amplicon sequencing. S. Enteritidis infection increased the abundance of Akkermansia and triggered the host inflammatory response. Conversely, the addition of flaxseed to the feed increased the abundance of beneficial intestinal bacteria, such as Lactobacilli and Bacteroides. Ovarian health is important for egg production performance in laying hens and our findings indicate that S. Enteritidis can persist in the ovaries for an extended period. Therefore, we further performed transcriptome sequencing analysis of ovarian tissues on day seven after S. Enteritidis infection. S. Enteritidis infection leads to altered ovarian gene expression, including the downregulation of lipid metabolism and growth and development genes and the upregulation of host immune response genes in laying hens. The upregulation of genes associated with growth and development may have stimulated ovarian growth and development.
Collapse
Affiliation(s)
- De Wang
- College of Life Sciences, Sichuan University, Chengdu 610044, China; (D.W.); (B.M.); (Z.L.); (W.L.); (T.Z.)
- Key Laboratory of Bio-Resource and Eco-Environment of the Ministry of Education, Chengdu 610064, China
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Boheng Ma
- College of Life Sciences, Sichuan University, Chengdu 610044, China; (D.W.); (B.M.); (Z.L.); (W.L.); (T.Z.)
- Key Laboratory of Bio-Resource and Eco-Environment of the Ministry of Education, Chengdu 610064, China
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Ziwei Liao
- College of Life Sciences, Sichuan University, Chengdu 610044, China; (D.W.); (B.M.); (Z.L.); (W.L.); (T.Z.)
- Key Laboratory of Bio-Resource and Eco-Environment of the Ministry of Education, Chengdu 610064, China
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Wenjing Li
- College of Life Sciences, Sichuan University, Chengdu 610044, China; (D.W.); (B.M.); (Z.L.); (W.L.); (T.Z.)
- Key Laboratory of Bio-Resource and Eco-Environment of the Ministry of Education, Chengdu 610064, China
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Tiejun Zhang
- College of Life Sciences, Sichuan University, Chengdu 610044, China; (D.W.); (B.M.); (Z.L.); (W.L.); (T.Z.)
- Key Laboratory of Bio-Resource and Eco-Environment of the Ministry of Education, Chengdu 610064, China
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Changwei Lei
- College of Life Sciences, Sichuan University, Chengdu 610044, China; (D.W.); (B.M.); (Z.L.); (W.L.); (T.Z.)
- Key Laboratory of Bio-Resource and Eco-Environment of the Ministry of Education, Chengdu 610064, China
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Hongning Wang
- College of Life Sciences, Sichuan University, Chengdu 610044, China; (D.W.); (B.M.); (Z.L.); (W.L.); (T.Z.)
- Key Laboratory of Bio-Resource and Eco-Environment of the Ministry of Education, Chengdu 610064, China
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| |
Collapse
|
5
|
Zhang F, Wang D. Potential of Akkermansia muciniphila and its outer membrane proteins as therapeutic targets for neuropsychological diseases. Front Microbiol 2023; 14:1191445. [PMID: 37440890 PMCID: PMC10333588 DOI: 10.3389/fmicb.2023.1191445] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/25/2023] [Indexed: 07/15/2023] Open
Abstract
The gut microbiota varies dramatically among individuals, and changes over time within the same individual, due to diversities in genetic backgrounds, diet, nutrient supplementations and use of antibiotics. Up until now, studies on dysbiosis of microbiota have expanded to a wider range of diseases, with Akkermansia muciniphila at the cross spot of many of these diseases. A. muciniphila is a Gram-negative bacterium that produces short-chain fatty acids (SCFAs), and Amuc_1100 is one of its most highly expressed outer membrane proteins. This review aims to summarize current knowledge on correlations between A. muciniphila and involved neuropsychological diseases published in the last decade, with a focus on the potential of this bacterium and its outer membrane proteins as therapeutic targets for these diseases, on the basis of evidence accumulated from animal and clinical studies, as well as mechanisms of action from peripheral to central nervous system (CNS).
Collapse
Affiliation(s)
- Fenghua Zhang
- Department of Laboratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Dali Wang
- Center for Clinical and Translational Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
6
|
Zhang L, Liu Y, Wang X, Zhang X. Physical Exercise and Diet: Regulation of Gut Microbiota to Prevent and Treat Metabolic Disorders to Maintain Health. Nutrients 2023; 15:nu15061539. [PMID: 36986268 PMCID: PMC10054346 DOI: 10.3390/nu15061539] [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: 03/07/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Each person's body is host to a large number and variety of gut microbiota, which has been described as the second genome and plays an important role in the body's metabolic process and is closely related to health. It is common knowledge that proper physical activity and the right diet structure can keep us healthy, and in recent years, researchers have found that this boost to health may be related to the gut microbiota. Past studies have reported that physical activity and diet can modulate the compositional structure of the gut microbiota and further influence the production of key metabolites of the gut microbiota, which can be an effective way to improve body metabolism and prevent and treat related metabolic diseases. In this review, we outline the role of physical activity and diet in regulating gut microbiota and the key role that gut microbiota plays in improving metabolic disorders. In addition, we highlight the regulation of gut microbiota through appropriate physical exercise and diet to improve body metabolism and prevent metabolic diseases, aiming to promote public health and provide a new approach to treating such diseases.
Collapse
Affiliation(s)
- Li Zhang
- Department of Physical Education, China University of Mining and Technology, Beijing 100083, China
| | - Yuan Liu
- Department of Physical Education, China University of Mining and Technology, Beijing 100083, China
| | - Xinzhou Wang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| |
Collapse
|
7
|
Ren T, Liu F, Wang D, Li B, Jiang P, Li J, Li H, Chen C, Wu W, Jiao L. Rhamnogalacturonan-I enriched pectin from steamed ginseng ameliorates lipid metabolism in type 2 diabetic rats via gut microbiota and AMPK pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 301:115862. [PMID: 36283638 DOI: 10.1016/j.jep.2022.115862] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/09/2022] [Accepted: 10/19/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax ginseng C. A. Meyer (Ginseng) has traditionally been used to treat diabetes. Polysaccharide is the main active component of ginseng, and has been proved to have hypoglycaemic and hypolipidaemic effects, but its mechanism remains unclear. AIM OF THE STUDY This study aimed to evaluate the effect and the potential mechanism of rhamnogalacturonan-I enriched pectin (GPS-1) from steamed ginseng on lipid metabolism in type 2 diabetes mellitus (T2DM) rats. MATERIALS AND METHODS GPS-1 was prepared by water extraction, ion-exchange and gel chromatography. High-glucose/high-fat diet combined with streptozotocin was used to establish T2DM rat models, and lipid levels in serum and liver were tested. 16S rRNA sequencing and gas chromatography-mass spectrometry were used to detect the changes of gut microbiota and metabolites. The protein and mRNA levels of lipid synthesis-related genes were detected by Western blot and quantitative real-time polymerase chain reaction. RESULTS The polyphagia, polydipsia, weight loss, hyperglycaemia, hyperlipidaemia and hepatic lipid accumulation in T2DM rats were alleviated after GPS-1 intervention. GPS-1 modulated the gut microbiota composition of T2DM rats, increased the levels of short-chain fatty acids, and promoted the secretion of glucagon-like peptide-1 and peptide tyrosine tyrosine. Further, GPS-1 activated AMP-activated protein kinases, phosphorylated acetyl-CoA carboxylase, reduced the expression of sterol regulatory element-binding protein-1c and fatty acid synthases in T2DM rats. CONCLUSIONS The regulation effects of GPS-1 on lipid metabolism in T2DM rats are related to the regulation of gut microbiota and activation of AMP-activated protein kinase pathway.
Collapse
Affiliation(s)
- Ting Ren
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Furao Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Dongxue Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Bo Li
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Peng Jiang
- National Demonstration Center for Experimental Biology Education, Northeast Normal University, Changchun, 130024, China.
| | - Junming Li
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Hui Li
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Changbao Chen
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Wei Wu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Lili Jiao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China.
| |
Collapse
|
8
|
May KS, den Hartigh LJ. Gut Microbial-Derived Short Chain Fatty Acids: Impact on Adipose Tissue Physiology. Nutrients 2023; 15:272. [PMID: 36678142 PMCID: PMC9865590 DOI: 10.3390/nu15020272] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Obesity is a global public health issue and major risk factor for pathological conditions, including type 2 diabetes, dyslipidemia, coronary artery disease, hepatic steatosis, and certain types of cancer. These metabolic complications result from a combination of genetics and environmental influences, thus contributing to impact whole-body homeostasis. Mechanistic animal and human studies have indicated that an altered gut microbiota can mediate the development of obesity, leading to inflammation beyond the intestine. Moreover, prior research suggests an interaction between gut microbiota and peripheral organs such as adipose tissue via different signaling pathways; yet, to what degree and in exactly what ways this inter-organ crosstalk modulates obesity remains elusive. This review emphasizes the influence of circulating gut-derived short chain fatty acids (SCFAs) i.e., acetate, propionate, and butyrate, on adipose tissue metabolism in the scope of obesity, with an emphasis on adipocyte physiology in vitro and in vivo. Furthermore, we discuss some of the well-established mechanisms via which microbial SCFAs exert a role as a prominent host energy source, hence regulating overall energy balance and health. Collectively, exploring the mechanisms via which SCFAs impact adipose tissue metabolism appears to be a promising avenue to improve metabolic conditions related to obesity.
Collapse
Affiliation(s)
- Karolline S. May
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA 98109, USA
- UW Medicine Diabetes Institute, 750 Republican Street, Box 358062, Seattle, WA 98109, USA
| | - Laura J. den Hartigh
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA 98109, USA
- UW Medicine Diabetes Institute, 750 Republican Street, Box 358062, Seattle, WA 98109, USA
| |
Collapse
|
9
|
Miyamoto J, Shimizu H, Hisa K, Matsuzaki C, Inuki S, Ando Y, Nishida A, Izumi A, Yamano M, Ushiroda C, Irie J, Katayama T, Ohno H, Itoh H, Yamamoto K, Kimura I. Host metabolic benefits of prebiotic exopolysaccharides produced by Leuconostoc mesenteroides. Gut Microbes 2023; 15:2161271. [PMID: 36604628 PMCID: PMC9828693 DOI: 10.1080/19490976.2022.2161271] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Fermented foods demonstrate remarkable health benefits owing to probiotic bacteria or microproducts produced via bacterial fermentation. Fermented foods are produced by the fermentative action of several lactic acid bacteria, including Leuconostoc mesenteroides; however, the exact mechanism of action of these foods remains unclear. Here, we observed that prebiotics associated with L. mesenteroides-produced exopolysaccharides (EPS) demonstrate substantial host metabolic benefits. L. mesenteroides-produced EPS is an indigestible α-glucan, and intake of the purified form of EPS improved glucose metabolism and energy homeostasis through EPS-derived gut microbial short-chain fatty acids, and changed gut microbial composition. Our findings reveal an important mechanism that accounts for the effects of diet, prebiotics, and probiotics on energy homeostasis and suggests an approach for preventing lifestyle-related diseases by targeting bacterial EPS.
Collapse
Affiliation(s)
- Junki Miyamoto
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Japan,AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Japan
| | - Hidenori Shimizu
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Japan,Noster Inc. Kamiueno, Muko-shi, Kyoto, Japan
| | - Keiko Hisa
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Japan,Noster Inc. Kamiueno, Muko-shi, Kyoto, Japan
| | - Chiaki Matsuzaki
- Research Institute for Bioresources and Biotechnology, Ishikawa Prefectural University, Ishikawa, Japan
| | - Shinsuke Inuki
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Japan,Department of Bioorganic Medicinal Chemistry and Chemogenomics, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Yuna Ando
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Akari Nishida
- Department of Molecular Neurobiology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Ayano Izumi
- Department of Molecular Neurobiology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Mayu Yamano
- Department of Molecular Neurobiology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Chihiro Ushiroda
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Japan,Department of Clinical Nutrition, Fujita Health University, Aichi, Japan
| | - Junichiro Irie
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Japan,Department of Endocrinology, Metabolism and Nephrology, School of Medicine, Keio University, Shinjuku-ku, Japan
| | - Takane Katayama
- Laboratory of Molecular Biology and Bioresponse, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Hiroaki Ohno
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Japan,Department of Bioorganic Medicinal Chemistry and Chemogenomics, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Hiroshi Itoh
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Japan,Department of Endocrinology, Metabolism and Nephrology, School of Medicine, Keio University, Shinjuku-ku, Japan
| | - Kenji Yamamoto
- Center for Innovative and Joint Research, Wakayama University, Wakayama, Japan
| | - Ikuo Kimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Japan,AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Japan,Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan,Department of Molecular Neurobiology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan,CONTACT Ikuo Kimura Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501183-8509, Japan
| |
Collapse
|
10
|
Ilyés T, Silaghi CN, Crăciun AM. Diet-Related Changes of Short-Chain Fatty Acids in Blood and Feces in Obesity and Metabolic Syndrome. BIOLOGY 2022; 11:1556. [PMID: 36358258 PMCID: PMC9687917 DOI: 10.3390/biology11111556] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 09/13/2023]
Abstract
Obesity-related illnesses are one of the leading causes of death worldwide. Metabolic syndrome has been associated with numerous health issues. Short-chain fatty acids (SCFAs) have been shown to have multiple effects throughout the body, both directly as well as through specific G protein-coupled receptors. The main SCFAs produced by the gut microbiota are acetate, propionate, and butyrate, which are absorbed in varying degrees from the large intestine, with some acting mainly locally and others systemically. Diet has the potential to influence the gut microbial composition, as well as the type and amount of SCFAs produced. High fiber-containing foods and supplements increase the production of SCFAs and SCFA-producing bacteria in the gut and have been shown to have bodyweight-lowering effects. Dietary supplements, which increase SCFA production, could open the way for novel approaches to weight loss interventions. The aim of this review is to analyze the variations of fecal and blood SCFAs in obesity and metabolic syndrome through a systematic search and analysis of existing literature.
Collapse
Affiliation(s)
| | - Ciprian N. Silaghi
- Department of Molecular Sciences, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400012 Cluj-Napoca, Romania
| | | |
Collapse
|
11
|
Wang L, Wang S, Zhang Q, He C, Fu C, Wei Q. The role of the gut microbiota in health and cardiovascular diseases. MOLECULAR BIOMEDICINE 2022; 3:30. [PMID: 36219347 PMCID: PMC9554112 DOI: 10.1186/s43556-022-00091-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/05/2022] [Indexed: 11/17/2022] Open
Abstract
The gut microbiota is critical to human health, such as digesting nutrients, forming the intestinal epithelial barrier, regulating immune function, producing vitamins and hormones, and producing metabolites to interact with the host. Meanwhile, increasing evidence indicates that the gut microbiota has a strong correlation with the occurrence, progression and treatment of cardiovascular diseases (CVDs). In patients with CVDs and corresponding risk factors, the composition and ratio of gut microbiota have significant differences compared with their healthy counterparts. Therefore, gut microbiota dysbiosis, gut microbiota-generated metabolites, and the related signaling pathway may serve as explanations for some of the mechanisms about the occurrence and development of CVDs. Several studies have also demonstrated that many traditional and latest therapeutic treatments of CVDs are associated with the gut microbiota and its generated metabolites and related signaling pathways. Given that information, we summarized the latest advances in the current research regarding the effect of gut microbiota on health, the main cardiovascular risk factors, and CVDs, highlighted the roles and mechanisms of several metabolites, and introduced corresponding promising treatments for CVDs regarding the gut microbiota. Therefore, this review mainly focuses on exploring the role of gut microbiota related metabolites and their therapeutic potential in CVDs, which may eventually provide better solutions in the development of therapeutic treatment as well as the prevention of CVDs.
Collapse
Affiliation(s)
- Lu Wang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Shiqi Wang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Qing Zhang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Chengqi He
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Chenying Fu
- grid.412901.f0000 0004 1770 1022National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,grid.412901.f0000 0004 1770 1022Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Quan Wei
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| |
Collapse
|
12
|
Zhang D, Liu J, Cheng H, Wang H, Tan Y, Feng W, Peng C. Interactions between polysaccharides and gut microbiota: A metabolomic and microbial review. Food Res Int 2022; 160:111653. [DOI: 10.1016/j.foodres.2022.111653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 12/17/2022]
|
13
|
Zhang L, Wang X, Zhang X. Modulation of Intestinal Flora by Dietary Polysaccharides: A Novel Approach for the Treatment and Prevention of Metabolic Disorders. Foods 2022; 11:2961. [PMID: 36230037 PMCID: PMC9562892 DOI: 10.3390/foods11192961] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/09/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
Intestinal flora is numerous and diverse, and play a key role in maintaining human health. Dietary polysaccharides are widely present in the daily diet and have a moderating effect on the intestinal flora. Past studies have confirmed that intestinal flora is involved in the metabolic process in the human body, and the change in intestinal flora structure is closely related to the metabolic disorders in the human body. Therefore, regulating intestinal flora through dietary polysaccharides is an effective way to treat and prevent common metabolic diseases and has great research value. However, this area has not received enough attention. In this review, we provide an overview of the modulatory effects of dietary polysaccharides on intestinal flora and the key role of intestinal flora in improving metabolic disorders in humans. In addition, we highlight the therapeutic and preventive effects of intestinal flora modulation through dietary polysaccharides on metabolic disorders, aiming to find new ways to treat metabolic disorders and facilitate future exploration in this field.
Collapse
Affiliation(s)
- Li Zhang
- Department of Physical Education, China University of Mining and Technology, Beijing 100083, China
| | - Xinzhou Wang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| |
Collapse
|
14
|
Kleigrewe K, Haack M, Baudin M, Ménabréaz T, Crovadore J, Masri M, Beyrer M, Andlauer W, Lefort F, Dawid C, Brück TB, Brück WM. Dietary Modulation of the Human Gut Microbiota and Metabolome with Flaxseed Preparations. Int J Mol Sci 2022; 23:ijms231810473. [PMID: 36142393 PMCID: PMC9499670 DOI: 10.3390/ijms231810473] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Flaxseeds are typically consumed either as whole flaxseed, ground flaxseed, flaxseed oil, partially defatted flaxseed meal, or as a milk alternative. They are considered a rich source of vitamins, minerals, proteins and peptides, lipids, carbohydrates, lignans, and dietary fiber, which have shown hypolipidemic, antiatherogenic, anticholesterolemic, and anti-inflammatory property activity. Here, an in vitro batch culture model was used to investigate the influence of whole milled flaxseed and partially defatted milled flaxseed press cake on the gut microbiota and the liberation of flaxseed bioactives. Microbial communities were profiled using 16S rRNA gene-based high-throughput sequencing with targeted mass spectrometry measuring lignan, cyclolinopeptide, and bile acid content and HPLC for short-chain fatty acid profiles. Flaxseed supplementation decreased gut microbiota richness with Firmicutes, Proteobacteria, and Bacteroidetes becoming the predominant phyla. Secoisolariciresinol, enterodiol, and enterolactone were rapidly produced with acetic acid, butyric acid, and propionic acid being the predominant acids after 24 h of fermentation. The flaxseed press cake and whole flaxseed were equivalent in microbiota changes and functionality. However, press cake may be superior as a functional additive in a variety of foods in terms of consumer acceptance as it would be more resistant to oxidative changes.
Collapse
Affiliation(s)
- Karin Kleigrewe
- Bavarian Center for Biomolecular Mass Spectrometry, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Martina Haack
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich, Garching b., 85748 München, Germany
| | - Martine Baudin
- Institute of Life Technologies, School of Engineering, HES-SO University of Applied Sciences and Arts Western Switzerland Valais-Wallis, 1950 Sion, Switzerland
| | - Thomas Ménabréaz
- Institute of Life Technologies, School of Engineering, HES-SO University of Applied Sciences and Arts Western Switzerland Valais-Wallis, 1950 Sion, Switzerland
| | - Julien Crovadore
- Plants and Pathogens Group, Research Institute Land Nature and Environment, Geneva School of Engineering, Architecture and Landscape (HEPIA), HES-SO University of Applied Sciences and Arts Western Switzerland, 1254 Jussy, Switzerland
| | - Mahmoud Masri
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich, Garching b., 85748 München, Germany
| | - Michael Beyrer
- Institute of Life Technologies, School of Engineering, HES-SO University of Applied Sciences and Arts Western Switzerland Valais-Wallis, 1950 Sion, Switzerland
| | - Wilfried Andlauer
- Institute of Life Technologies, School of Engineering, HES-SO University of Applied Sciences and Arts Western Switzerland Valais-Wallis, 1950 Sion, Switzerland
| | - François Lefort
- Plants and Pathogens Group, Research Institute Land Nature and Environment, Geneva School of Engineering, Architecture and Landscape (HEPIA), HES-SO University of Applied Sciences and Arts Western Switzerland, 1254 Jussy, Switzerland
| | - Corinna Dawid
- Bavarian Center for Biomolecular Mass Spectrometry, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
- Chair of Food Chemistry and Molecular Sensory Science, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Thomas B. Brück
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich, Garching b., 85748 München, Germany
| | - Wolfram M. Brück
- Institute of Life Technologies, School of Engineering, HES-SO University of Applied Sciences and Arts Western Switzerland Valais-Wallis, 1950 Sion, Switzerland
- Correspondence: ; Tel.: +41-58-606-86-64
| |
Collapse
|
15
|
Ribeiro FM, Silva MA, Lyssa V, Marques G, Lima HK, Franco OL, Petriz B. The molecular signaling of exercise and obesity in the microbiota-gut-brain axis. Front Endocrinol (Lausanne) 2022; 13:927170. [PMID: 35966101 PMCID: PMC9365995 DOI: 10.3389/fendo.2022.927170] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity is one of the major pandemics of the 21st century. Due to its multifactorial etiology, its treatment requires several actions, including dietary intervention and physical exercise. Excessive fat accumulation leads to several health problems involving alteration in the gut-microbiota-brain axis. This axis is characterized by multiple biological systems generating a network that allows bidirectional communication between intestinal bacteria and brain. This mutual communication maintains the homeostasis of the gastrointestinal, central nervous and microbial systems of animals. Moreover, this axis involves inflammatory, neural, and endocrine mechanisms, contributes to obesity pathogenesis. The axis also acts in appetite and satiety control and synthesizing hormones that participate in gastrointestinal functions. Exercise is a nonpharmacologic agent commonly used to prevent and treat obesity and other chronic degenerative diseases. Besides increasing energy expenditure, exercise induces the synthesis and liberation of several muscle-derived myokines and neuroendocrine peptides such as neuropeptide Y, peptide YY, ghrelin, and leptin, which act directly on the gut-microbiota-brain axis. Thus, exercise may serve as a rebalancing agent of the gut-microbiota-brain axis under the stimulus of chronic low-grade inflammation induced by obesity. So far, there is little evidence of modification of the gut-brain axis as a whole, and this narrative review aims to address the molecular pathways through which exercise may act in the context of disorders of the gut-brain axis due to obesity.
Collapse
Affiliation(s)
- Filipe M. Ribeiro
- Post-Graduation Program in Physical Education, Catholic University of Brasilia, Brasilia, Brazil
- Center for Proteomic and Biochemical Analysis, Post-Graduation in Genomic and Biotechnology Sciences, Catholic University of Brasilia, Brasília, Brazil
- Laboratory of Molecular Exercise Physiology - University Center of the Federal District - UDF, Brasilia, Brazil
| | - Maycon A. Silva
- Center for Proteomic and Biochemical Analysis, Post-Graduation in Genomic and Biotechnology Sciences, Catholic University of Brasilia, Brasília, Brazil
| | - Victória Lyssa
- Laboratory of Molecular Analysis, Graduate Program of Sciences and Technology of Health, University of Brasilia, Brasilia, Brazil
| | - Gabriel Marques
- Laboratory of Molecular Exercise Physiology - University Center of the Federal District - UDF, Brasilia, Brazil
| | - Henny K. Lima
- Center for Proteomic and Biochemical Analysis, Post-Graduation in Genomic and Biotechnology Sciences, Catholic University of Brasilia, Brasília, Brazil
| | - Octavio L. Franco
- Post-Graduation Program in Physical Education, Catholic University of Brasilia, Brasilia, Brazil
- Center for Proteomic and Biochemical Analysis, Post-Graduation in Genomic and Biotechnology Sciences, Catholic University of Brasilia, Brasília, Brazil
- S-Inova Biotech, Catholic University Dom Bosco, Biotechnology Program, Campo Grande, Brazil
| | - Bernardo Petriz
- Center for Proteomic and Biochemical Analysis, Post-Graduation in Genomic and Biotechnology Sciences, Catholic University of Brasilia, Brasília, Brazil
- Laboratory of Molecular Exercise Physiology - University Center of the Federal District - UDF, Brasilia, Brazil
- Postgraduate Program in Rehabilitation Sciences - University of Brasília, Brasília, Brazil
| |
Collapse
|
16
|
Dantas Machado AC, Brown SD, Lingaraju A, Sivaganesh V, Martino C, Chaix A, Zhao P, Pinto AFM, Chang MW, Richter RA, Saghatelian A, Saltiel AR, Knight R, Panda S, Zarrinpar A. Diet and feeding pattern modulate diurnal dynamics of the ileal microbiome and transcriptome. Cell Rep 2022; 40:111008. [PMID: 35793637 PMCID: PMC9296000 DOI: 10.1016/j.celrep.2022.111008] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 03/28/2022] [Accepted: 06/03/2022] [Indexed: 12/30/2022] Open
Abstract
Compositional oscillations of the gut microbiome are essential for normal peripheral circadian rhythms, both of which are disrupted in diet-induced obesity (DIO). Although time-restricted feeding (TRF) maintains circadian synchrony and protects against DIO, its impact on the dynamics of the cecal gut microbiome is modest. Thus, other regions of the gut, particularly the ileum, the nexus for incretin and bile acid signaling, may play an important role in entraining peripheral circadian rhythms. We demonstrate the effect of diet and feeding rhythms on the ileal microbiome composition and transcriptome in mice. The dynamic rhythms of ileal microbiome composition and transcriptome are dampened in DIO. TRF partially restores diurnal rhythms of the ileal microbiome and transcriptome, increases GLP-1 release, and alters the ileal bile acid pool and farnesoid X receptor (FXR) signaling, which could explain how TRF exerts its metabolic benefits. Finally, we provide a web resource for exploration of ileal microbiome and transcriptome circadian data.
Collapse
Affiliation(s)
- Ana Carolina Dantas Machado
- Division of Gastroenterology, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA
| | - Steven D Brown
- Division of Gastroenterology, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA
| | - Amulya Lingaraju
- Division of Gastroenterology, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA
| | - Vignesh Sivaganesh
- Division of Gastroenterology, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA
| | - Cameron Martino
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA, USA; Center for Microbiome Innovation, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA
| | - Amandine Chaix
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Peng Zhao
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Antonio F M Pinto
- Clayton Foundation Laboratories for Peptide Biology, the Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Max W Chang
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA
| | - R Alexander Richter
- Division of Gastroenterology, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, the Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Alan R Saltiel
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Pharmacology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA; Institute of Diabetes and Metabolic Health, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA
| | - Rob Knight
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Center for Microbiome Innovation, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Satchidananda Panda
- Regulatory Biology Laboratory, The Salk Institute, 10010 N. Torrey Pines Road, La Jolla, CA, USA
| | - Amir Zarrinpar
- Division of Gastroenterology, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Center for Microbiome Innovation, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Institute of Diabetes and Metabolic Health, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; VA Health Sciences, San Diego, La Jolla, CA, USA.
| |
Collapse
|
17
|
Takei N, Kuda T, Handa N, Fujita S, Takahashi H, Kimura B. Detection and isolation of typical gut indigenous bacteria in ICR mice fed wheat bran and wheat straw fibre. FOOD CHEMISTRY: MOLECULAR SCIENCES 2022; 4:100071. [PMID: 35415683 PMCID: PMC8991949 DOI: 10.1016/j.fochms.2021.100071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/10/2021] [Accepted: 12/28/2021] [Indexed: 11/30/2022]
Abstract
Wheat bran (WB) and wheat straw fibre (WSF) were administered ICR mice for 14 days. Caecal microbiota was determined by 16S rDNA amplicon sequencing. Caecal Lactobacillus johnsonii and Limosilactobacillus reuteri were high in WB diet-fed mice. L. johnsonii Wheat-1 and L. reuteri Wheat-12 strains could be isolated. WB fermentation with L. johnsonii Wheat-1 induce superoxide anion radical scavenging capacity.
Wheat bran (WB) and wheat straw fibre (WSF) are by-products of the wheat flour industry. To prove the existence of indigenous gut bacteria responsible for WB and WSF, the Institute of Cancer Research (ICR) mice were fed a diet containing no fibre (CS), 10% WB, or 5% WSF for 14 d. The caecal microbiome was analysed by 16S rDNA (V4 region) amplicon sequencing. Typical colonies were isolated and estimated by 16S rRNA gene BLASTn analysis. The predominant amplicon sequence variants in all diet groups belonged to Bifidobacterium pseudolongum- and Faecalibaculum rodentium-like bacteria. Lactobacillus johnsonii- and Limosilactobacillus reuteri-like bacteria were high in the WB group compared with those in the CS group. Lactobacillus johnsonii Wheat-1 and L. reuteri Wheat-12 strains could be isolated. Lactobacillus johnsonii Wheat-1 exhibited good fermentation activity in 10% (w/v) WB suspension. Superoxide anion radical scavenging capacity of the WB suspension was significantly increased by the fermentation.
Collapse
|
18
|
You H, Tan Y, Yu D, Qiu S, Bai Y, He J, Cao H, Che Q, Guo J, Su Z. The Therapeutic Effect of SCFA-Mediated Regulation of the Intestinal Environment on Obesity. Front Nutr 2022; 9:886902. [PMID: 35662937 PMCID: PMC9157426 DOI: 10.3389/fnut.2022.886902] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/20/2022] [Indexed: 12/12/2022] Open
Abstract
Intestinal environment disorder is a potential pathological mechanism of obesity. There is increasing evidence that disorders in the homeostasis of the intestinal environment can affect various metabolic organs, such as fat and liver, and lead to metabolic diseases. However, there are few therapeutic approaches for obesity targeting the intestinal environment. In this review, on the one hand, we discuss how intestinal microbial metabolites SCFA regulate intestinal function to improve obesity and the possible mechanisms and pathways related to obesity-related pathological processes (depending on SCFA-related receptors such as GPCRs, MCT and SMCT, and through epigenetic processes). On the other hand, we discuss dietary management strategies to enrich SCFA-producing bacteria and target specific SCFA-producing bacteria and whether fecal bacteria transplantation therapy to restore the composition of the gut microbiota to regulate SCFA can help prevent or improve obesity. Finally, we believe that it will be of great significance to establish a working model of gut– SCFA– metabolic disease development in the future for the improvement this human health concern.
Collapse
Affiliation(s)
- Huimin You
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, China.,Key Laboratory of Glucolipid Metabolic Disorder, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Ministry of Education of China, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yue Tan
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, China.,Key Laboratory of Glucolipid Metabolic Disorder, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Ministry of Education of China, Guangdong Pharmaceutical University, Guangzhou, China
| | - Dawei Yu
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, China.,Key Laboratory of Glucolipid Metabolic Disorder, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Ministry of Education of China, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shuting Qiu
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, China.,Key Laboratory of Glucolipid Metabolic Disorder, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Ministry of Education of China, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jincan He
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hua Cao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qishi Che
- Guangzhou Rainhome Pharm & Tech Co., Ltd, Guangzhou, China
| | - Jiao Guo
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Ministry of Education of China, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, China.,Key Laboratory of Glucolipid Metabolic Disorder, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Ministry of Education of China, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
19
|
Smith C, Van Haute MJ, Xian Y, Segura Munoz RR, Liu S, Schmaltz RJ, Ramer-Tait AE, Rose DJ. Carbohydrate utilization by the gut microbiome determines host health responsiveness to whole grain type and processing methods. Gut Microbes 2022; 14:2126275. [PMID: 36130094 PMCID: PMC9519025 DOI: 10.1080/19490976.2022.2126275] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/02/2022] [Accepted: 09/13/2022] [Indexed: 02/04/2023] Open
Abstract
Little is known about how interactions among grain processing, grain type, and carbohydrate utilization (CU) by the microbiome influence the health benefits of whole grains. Therefore, two whole grains - brown rice and whole wheat - and two processing methods - boiling (porridge) and extrusion - were studied for their effects on host metabolic outcomes in mice harboring human microbiomes previously shown in vitro to have high or low CU. Mice carrying either microbiome experienced increases in body weight and glycemia when consuming Western diets supplemented with extruded grains versus porridge. However, mice with the high but not low CU microbiome also gained more weight and fat over time and were less glucose tolerant when consuming extruded grain diets. In high CU microbiome mice, the exacerbated negative health outcomes associated with extrusion were related to altered abundances of Lachnospiraceae and Ruminococcaceae as well as elevated sugar degradation and colonic acetate production. The amplicon sequence variants (ASVs) associated with extruded and porridge diets in this in vivo study were not the same as those identified in our prior in vitro study; however, the predicted functions were highly correlated. In conclusion, mice harboring both high and low CU microbiomes responded to the whole grain diets similarly, except the high CU microbiome mice exhibited exacerbated effects due to excessive acetate production, indicating that CU by the microbiome is linked to host metabolic health outcomes. Our work demonstrates that a greater understanding of food processing effects on the microbiome is necessary for developing foods that promote rather than diminish host health.Abbreviations: CU- carbohydrate utilization; SCFA- short-chain fatty acids; GF- germ-free; HMA, human-microbiome associated; ipGTT- intraperitoneal glucose tolerance test; HOMA-IR- Homeostatic Model Assessment for Insulin Resistance; AUC- area under the glycemia curve; ASV- amplicon sequence variant; lf- low-fat; wd- Western diet; wd_wwp- Western diet containing whole wheat porridge; wd_wwe- Western diet containing whole wheat extrudate; wd_bre- Western diet containing brown rice extrudate; wd_extr- Western diet containing either whole wheat or brown rice extrudate.
Collapse
Affiliation(s)
- Caroline Smith
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
- Grain Research and Quality, Ardent Mills, Denver, CO, USA
| | - Mallory J. Van Haute
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
- Research and Development, Synbiotic Health, Lincoln, NE, USA
| | - Yibo Xian
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Rafael R. Segura Munoz
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
- Department of Systems & Synthetic Microbiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Sujun Liu
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Robert J. Schmaltz
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Amanda E. Ramer-Tait
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Devin J. Rose
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
- Department of Agronomy and Horticulture, University of Nebraska-Lincoln, Lincoln, NE, USA
| |
Collapse
|
20
|
Dietary Phytoestrogens and Their Metabolites as Epigenetic Modulators with Impact on Human Health. Antioxidants (Basel) 2021; 10:antiox10121893. [PMID: 34942997 PMCID: PMC8750933 DOI: 10.3390/antiox10121893] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/17/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
The impact of dietary phytoestrogens on human health has been a topic of continuous debate since their discovery. Nowadays, based on their presumptive beneficial effects, the amount of phytoestrogens consumed in the daily diet has increased considerably worldwide. Thus, there is a growing need for scientific data regarding their mode of action in the human body. Recently, new insights of phytoestrogens’ bioavailability and metabolism have demonstrated an inter-and intra-population heterogeneity of final metabolites’ production. In addition, the phytoestrogens may have the ability to modulate epigenetic mechanisms that control gene expression. This review highlights the complexity and particularity of the metabolism of each class of phytoestrogens, pointing out the diversity of their bioactive gut metabolites. Futhermore, it presents emerging scientific data which suggest that, among well-known genistein and resveratrol, other phytoestrogens and their gut metabolites can act as epigenetic modulators with a possible impact on human health. The interconnection of dietary phytoestrogens’ consumption with gut microbiota composition, epigenome and related preventive mechanisms is discussed. The current challenges and future perspectives in designing relevant research directions to explore the potential health benefits of dietary phytoestrogens are also explored.
Collapse
|
21
|
Modulation of Adipocyte Metabolism by Microbial Short-Chain Fatty Acids. Nutrients 2021; 13:nu13103666. [PMID: 34684670 PMCID: PMC8538331 DOI: 10.3390/nu13103666] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/17/2021] [Accepted: 10/17/2021] [Indexed: 12/13/2022] Open
Abstract
Obesity and its complications—including type 2 diabetes, cardiovascular disease, and certain cancers—constitute a rising global epidemic that has imposed a substantial burden on health and healthcare systems over the years. It is becoming increasingly clear that there is a link between obesity and the gut microbiota. Gut dysbiosis, characterized as microbial imbalance, has been consistently associated with obesity in both humans and animal models, and can be reversed with weight loss. Emerging evidence has shown that microbial-derived metabolites such as short-chain fatty acids (SCFAs)—including acetate, propionate, and butyrate—provide benefits to the host by impacting organs beyond the gut, including adipose tissue. In this review, we summarize what is currently known regarding the specific mechanisms that link gut-microbial-derived SCFAs with adipose tissue metabolism, such as adipogenesis, lipolysis, and inflammation. In addition, we explore indirect mechanisms by which SCFAs can modulate adipose tissue metabolism, such as via perturbation of gut hormones, as well as signaling to the brain and the liver. Understanding how the modulation of gut microbial metabolites such as SCFAs can impact adipose tissue function could lead to novel therapeutic strategies for the prevention and treatment of obesity.
Collapse
|
22
|
Arora T, Vanslette AM, Hjorth SA, Bäckhed F. Microbial regulation of enteroendocrine cells. MED 2021; 2:553-570. [DOI: 10.1016/j.medj.2021.03.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 02/08/2023]
|
23
|
Carey RA, Montag D. Exploring the relationship between gut microbiota and exercise: short-chain fatty acids and their role in metabolism. BMJ Open Sport Exerc Med 2021; 7:e000930. [PMID: 33981447 PMCID: PMC8061837 DOI: 10.1136/bmjsem-2020-000930] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2021] [Indexed: 12/16/2022] Open
Abstract
The human body is host to a multitude of bacteria, fungi, viruses and other species in the intestine, collectively known as the microbiota. Dietary carbohydrates which bypass digestion and absorption are broken down and fermented by the microbiota to produce short-chain fatty acids (SCFAs). Previous research has established the role of SCFAs in the control of human metabolic pathways. In this review, we evaluate SCFAs as a metabolic regulator and how they might improve endurance performance in athletes. By looking at research conducted in animal models, we identify several pathways downstream of SCFAs, either directly modulating metabolic pathways through second messenger pathways or through neuronal pathways, that contribute to energy utilisation. These pathways contribute to efficient energy metabolism and are thus key to maximising substrate utilisation in endurance exercise. Future research may prove the usefulness of targeted dietary interventions allowing athletes to maximise their performance in competition.
Collapse
Affiliation(s)
- Ryan A Carey
- Global Public Health, Queen Mary University of London, London, UK
| | - Doreen Montag
- Global Public Health, Queen Mary University of London, London, UK
| |
Collapse
|
24
|
Free Fatty Acid Receptors 2 and 3 as Microbial Metabolite Sensors to Shape Host Health: Pharmacophysiological View. Biomedicines 2020; 8:biomedicines8060154. [PMID: 32521775 PMCID: PMC7344995 DOI: 10.3390/biomedicines8060154] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/30/2020] [Accepted: 06/01/2020] [Indexed: 12/13/2022] Open
Abstract
The role of the gut microbiome in human health is becoming apparent. The major functional impact of the gut microbiome is transmitted through the microbial metabolites that are produced in the gut and interact with host cells either in the local gut environment or are absorbed into circulation to impact distant cells/organs. Short-chain fatty acids (SCFAs) are the major microbial metabolites that are produced in the gut through the fermentation of non-digestible fibers. SCFAs are known to function through various mechanisms, however, their signaling through free fatty acid receptors 2 and 3 (FFAR2/3; type of G-coupled protein receptors) is a new therapeutic approach. FFAR2/3 are widely expressed in diverse cell types in human and mice, and function as sensors of SCFAs to change several physiological and cellular functions. FFAR2/3 modulate neurological signaling, energy metabolism, intestinal cellular homeostasis, immune response, and hormone synthesis. FFAR2/3 function through Gi and/or Gq signaling, that is mediated through specific structural features of SCFAs-FFAR2/3 bindings and modulating specific signaling pathway. In this review, we discuss the wide-spread expression and structural homologies between human and mice FFAR2/3, and their role in different human health conditions. This information can unlock opportunities to weigh the potential of FFAR2/3 as a drug target to prevent human diseases.
Collapse
|
25
|
Di Luccia B, Ahern PP, Griffin NW, Cheng J, Guruge JL, Byrne AE, Rodionov DA, Leyn SA, Osterman AL, Ahmed T, Colonna M, Barratt MJ, Delahaye NF, Gordon JI. Combined Prebiotic and Microbial Intervention Improves Oral Cholera Vaccination Responses in a Mouse Model of Childhood Undernutrition. Cell Host Microbe 2020; 27:899-908.e5. [PMID: 32348782 DOI: 10.1016/j.chom.2020.04.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/18/2020] [Accepted: 04/08/2020] [Indexed: 12/11/2022]
Abstract
Undernourished children in low-income countries often exhibit poor responses to oral vaccination. Perturbed microbiota development is linked to undernutrition, but whether and how microbiota changes affect vaccine responsiveness remains unclear. Here, we show that gnotobiotic mice colonized with microbiota from undernourished Bangladeshi children and fed a Bangladeshi diet exhibited microbiota-dependent differences in mucosal IgA responses to oral vaccination with cholera toxin (CT). Supplementation with a nutraceutical consisting of spirulina, amaranth, flaxseed, and micronutrients augmented CT-IgA production. Mice initially colonized with a microbiota associated with poor CT responses exhibited improved immunogenicity upon invasion of bacterial taxa from cagemates colonized with a more "responsive" microbiota. Additionally, a consortium of five cultured bacterial invaders conferred augmented CT-IgA responses in mice fed the supplemented diet and colonized with the "hypo-responsive" community. These results provide preclinical proof-of-concept that diet and microbiota influence mucosal immune responses to CT vaccination and identify a candidate synbiotic formulation.
Collapse
Affiliation(s)
- Blanda Di Luccia
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Philip P Ahern
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nicholas W Griffin
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jiye Cheng
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Janaki L Guruge
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alexandra E Byrne
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dmitry A Rodionov
- A.A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow 127994, Russia; Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Semen A Leyn
- A.A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow 127994, Russia; Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Andrei L Osterman
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Tahmeed Ahmed
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael J Barratt
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Jeffrey I Gordon
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
26
|
Gut Microbiome: Profound Implications for Diet and Disease. Nutrients 2019; 11:nu11071613. [PMID: 31315227 PMCID: PMC6682904 DOI: 10.3390/nu11071613] [Citation(s) in RCA: 642] [Impact Index Per Article: 107.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/05/2019] [Accepted: 07/11/2019] [Indexed: 02/07/2023] Open
Abstract
The gut microbiome plays an important role in human health and influences the development of chronic diseases ranging from metabolic disease to gastrointestinal disorders and colorectal cancer. Of increasing prevalence in Western societies, these conditions carry a high burden of care. Dietary patterns and environmental factors have a profound effect on shaping gut microbiota in real time. Diverse populations of intestinal bacteria mediate their beneficial effects through the fermentation of dietary fiber to produce short-chain fatty acids, endogenous signals with important roles in lipid homeostasis and reducing inflammation. Recent progress shows that an individual’s starting microbial profile is a key determinant in predicting their response to intervention with live probiotics. The gut microbiota is complex and challenging to characterize. Enterotypes have been proposed using metrics such as alpha species diversity, the ratio of Firmicutes to Bacteroidetes phyla, and the relative abundance of beneficial genera (e.g., Bifidobacterium, Akkermansia) versus facultative anaerobes (E. coli), pro-inflammatory Ruminococcus, or nonbacterial microbes. Microbiota composition and relative populations of bacterial species are linked to physiologic health along different axes. We review the role of diet quality, carbohydrate intake, fermentable FODMAPs, and prebiotic fiber in maintaining healthy gut flora. The implications are discussed for various conditions including obesity, diabetes, irritable bowel syndrome, inflammatory bowel disease, depression, and cardiovascular disease.
Collapse
|
27
|
De Silva SF, Alcorn J. Flaxseed Lignans as Important Dietary Polyphenols for Cancer Prevention and Treatment: Chemistry, Pharmacokinetics, and Molecular Targets. Pharmaceuticals (Basel) 2019; 12:E68. [PMID: 31060335 PMCID: PMC6630319 DOI: 10.3390/ph12020068] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/26/2019] [Accepted: 04/30/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer causes considerable morbidity and mortality across the world. Socioeconomic, environmental, and lifestyle factors contribute to the increasing cancer prevalence, bespeaking a need for effective prevention and treatment strategies. Phytochemicals like plant polyphenols are generally considered to have anticancer, anti-inflammatory, antiviral, antimicrobial, and immunomodulatory effects, which explain their promotion for human health. The past several decades have contributed to a growing evidence base in the literature that demonstrate ability of polyphenols to modulate multiple targets of carcinogenesis linking models of cancer characteristics (i.e., hallmarks and nutraceutical-based targeting of cancer) via direct or indirect interaction or modulation of cellular and molecular targets. This evidence is particularly relevant for the lignans, an ubiquitous, important class of dietary polyphenols present in high levels in food sources such as flaxseed. Literature evidence on lignans suggests potential benefit in cancer prevention and treatment. This review summarizes the relevant chemical and pharmacokinetic properties of dietary polyphenols and specifically focuses on the biological targets of flaxseed lignans. The consolidation of the considerable body of data on the diverse targets of the lignans will aid continued research into their potential for use in combination with other cancer chemotherapies, utilizing flaxseed lignan-enriched natural products.
Collapse
Affiliation(s)
- S Franklyn De Silva
- Drug Discovery & Development Research Group, College of Pharmacy and Nutrition, 104 Clinic Place, Health Sciences Building, University of Saskatchewan, Saskatoon, Saskatchewan (SK), S7N 2Z4, Canada.
| | - Jane Alcorn
- Drug Discovery & Development Research Group, College of Pharmacy and Nutrition, 104 Clinic Place, Health Sciences Building, University of Saskatchewan, Saskatoon, Saskatchewan (SK), S7N 2Z4, Canada.
| |
Collapse
|