1
|
Mukhopadhya I, Louis P. Gut microbiota-derived short-chain fatty acids and their role in human health and disease. Nat Rev Microbiol 2025:10.1038/s41579-025-01183-w. [PMID: 40360779 DOI: 10.1038/s41579-025-01183-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2025] [Indexed: 05/15/2025]
Abstract
Short-chain fatty acids (SCFAs) are a group of organic compounds produced by the fermentation of dietary fibre by the human gut microbiota. They play diverse roles in different physiological processes of the host with implications for human health and disease. This Review provides an overview of the complex microbial metabolism underlying SCFA formation, considering microbial interactions and modulating factors of the gut environment. We explore the multifaceted mechanistic interactions between SCFAs and the host, with a particular focus on the local actions of SCFAs in the gut and their complex interactions with the immune system. We also discuss how these actions influence intestinal and extraintestinal diseases and emerging therapeutic strategies using SCFAs.
Collapse
Affiliation(s)
- Indrani Mukhopadhya
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Petra Louis
- Rowett Institute, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
2
|
van Deuren T, Umanets A, Venema K, Moreno LL, Zoetendal EG, Canfora EE, Blaak EE. Specific dietary fibers steer toward distal colonic saccharolytic fermentation using the microbiota of individuals with overweight/obesity. Food Res Int 2025; 209:116271. [PMID: 40253188 DOI: 10.1016/j.foodres.2025.116271] [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: 01/07/2025] [Revised: 02/28/2025] [Accepted: 03/11/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Evidence suggests that increased distal short-chain fatty acid (SCFA) production beneficially impacts metabolic health. However, indigestible carbohydrate availability is limited in the distal colon; consequently, microbes shift toward protein fermentation, often linked to adverse metabolic health effects. We aimed to identify specific fiber(s) that promote saccharolytic fermentation in the distal colon and thereby may (partially) inhibit proteolytic fermentation. METHODS Potato-fiber, pectin, and inulin were studied individually and in combination against a high (predigested) protein background using an in vitro model of the colon (TIM-2) inoculated with pooled, standardized fecal microbiota from individuals with overweight/obesity. Microbiota composition and activity were assessed at different timepoints to simulate the travel throughout the colon (proximal: 0-8 h, distal: 8-24 h) and compared to a high protein (HP)_control, receiving only proteins. RESULTS Fiber addition increased total SCFA production compared to HP_control (52.11 ± 1.49 vs 27.07 ± 0.26 mmol) whereas total branched-chain fatty acids (BCFA; a marker for protein fermentation) production only slightly decreased (3.31 ± 0.10 vs 4.18 ± 0.40 mmol). Combining potato-fiber and pectin led to the highest total and distal SCFA production and distal SCFA:BCFA. Fiber addition attenuated HP-induced increases in several bacterial taxa including Mogibacterium and Coprococcus, independent of fiber type. Additionally, time- and fiber-specific microbial signatures were identified: inulin increased Bifidobacterium (proximal) relative abundance and pectin and/or potato-fiber increased Prevotella 9 (distal) relative abundance. CONCLUSION The most marked increase in distal colonic SCFA production was induced by combining potato-fiber and pectin. Further research should elucidate whether this switch toward saccharolytic fermentation translates into beneficial metabolic health effects in humans.
Collapse
Affiliation(s)
- Thirza van Deuren
- Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, the Netherlands'
| | - Alexander Umanets
- Chair Group Youth Food and Health, Faculty of Science and Engineering, Maastricht University-Campus Venlo, Venlo, the Netherlands; Centre for Healthy Eating & Food Innovation (HEFI), Maastricht University-Campus Venlo, Venlo, the Netherlands
| | - Koen Venema
- Centre for Healthy Eating & Food Innovation (HEFI), Maastricht University-Campus Venlo, Venlo, the Netherlands
| | - Luis L Moreno
- Laboratory of Food Chemistry, Wageningen University and Research, Bornse Weilanden 9, 6708, WG, Wageningen, the Netherlands; Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708, WE, Wageningen, the Netherlands
| | - Erwin G Zoetendal
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708, WE, Wageningen, the Netherlands
| | - Emanuel E Canfora
- Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, the Netherlands'
| | - Ellen E Blaak
- Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, the Netherlands'.
| |
Collapse
|
3
|
Liu L, Qi W, Zhang N, Zhang J, Liu S, Wang H, Jiang L, Sun Y. Nutraceuticals for Gut-Brain Axis Health: A Novel Approach to Combat Malnutrition and Future Personalised Nutraceutical Interventions. Nutrients 2025; 17:1551. [PMID: 40362863 PMCID: PMC12073618 DOI: 10.3390/nu17091551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/22/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
The gut-brain axis (GBA) is a bidirectional communication network between the gastrointestinal tract and the brain, modulated by gut microbiota and related biomarkers. Malnutrition disrupts GBA homeostasis, exacerbating GBA dysfunction through gut dysbiosis, impaired neuroactive metabolite production, and systemic inflammation. Nutraceuticals, including probiotics, prebiotics, synbiotics, postbiotics, and paraprobiotics, offer a promising approach to improving GBA homeostasis by modulating the gut microbiota composition and related neuroactive metabolites. This review aims to elucidate the interplay between gut microbiota-derived biomarkers and GBA dysfunction in malnutrition and evaluate the potential of nutraceuticals in combating malnutrition. Furthermore, it explores the future of personalised nutraceutical interventions tailored to individual genetic and microbiome profiles, providing a targeted approach to optimise health outcomes. The integration of nutraceuticals into GBA health management could transform malnutrition treatment and improve cognitive and metabolic health.
Collapse
Affiliation(s)
- Litai Liu
- Tourism & Cuisine College, Harbin University of Commerce, Harbin 150028, China; (L.L.); (W.Q.); (N.Z.); (S.L.)
- Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6UR, UK
| | - Wen Qi
- Tourism & Cuisine College, Harbin University of Commerce, Harbin 150028, China; (L.L.); (W.Q.); (N.Z.); (S.L.)
| | - Na Zhang
- Tourism & Cuisine College, Harbin University of Commerce, Harbin 150028, China; (L.L.); (W.Q.); (N.Z.); (S.L.)
| | - Jinhao Zhang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.Z.); (H.W.); (L.J.)
| | - Shen Liu
- Tourism & Cuisine College, Harbin University of Commerce, Harbin 150028, China; (L.L.); (W.Q.); (N.Z.); (S.L.)
| | - Huan Wang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.Z.); (H.W.); (L.J.)
| | - Lianzhou Jiang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.Z.); (H.W.); (L.J.)
| | - Ying Sun
- Tourism & Cuisine College, Harbin University of Commerce, Harbin 150028, China; (L.L.); (W.Q.); (N.Z.); (S.L.)
| |
Collapse
|
4
|
Hou Y, Gao X, Gong J, Dong X, Hao Y, Zhai Z, Zhang H, Zhang M, Liu R, Wang R, Zhao L. Targeted Sodium Acetate Liposomes for Hepatocytes and Kupffer Cells: An Oral Dual-Targeted Therapeutic Approach for Non-Alcoholic Fatty Liver Disease Alleviation. Nutrients 2025; 17:930. [PMID: 40077800 PMCID: PMC11901740 DOI: 10.3390/nu17050930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: Sodium acetate (NaA) has demonstrated potential in improving non-alcoholic fatty liver disease (NAFLD) by targeting hepatocytes and Kupffer cells. However, its clinical application is hindered by low oral bioavailability and insufficient liver concentrations. Liposomes, with their capacity to encapsulate water-soluble drugs and be surface-modified, offer a promising solution for targeted oral drug delivery. Methods: We designed NaA-loaded liposomes modified with sodium cholate (SC) and mannose (MAN) (NaA@SC/MAN-LPs) to target hepatocytes and Kupffer cells. Results: The NaA@SC/MAN-LPs had a mean diameter of approximately 100 nm with a positive surface charge. Compared to free NaA, NaA@SC/MAN-LPs significantly extended the serum half-life from 2.85 h to 15.58 h, substantially improving in vivo bioavailability. In vivo distribution studies revealed that NaA@SC/MAN-LPs extended the acetate peak time in the liver from 15 min to 60 min and increased hepatic acetate accumulation to 3.75 times that of free NaA. In in vitro cell experiments, NaA@SC/MAN-LPs significantly reduced the lipid droplet, triglycerides (TG), and total cholesterol (TC) in a fatty acid-induced hepatocyte steatosis model and suppressed proinflammation in a lipopolysaccharide (LPS)-activated Kupffer cell inflammation model. Free NaA effectively improved hepatic lipid deposition in NAFLD mice. Furthermore, NaA@SC/MAN-LPs decreased hepatic TG, TC, and the relative area of lipid droplets by 30.44%, 15.26%, and 55.83%, compared to free NaA. Furthermore, the liposomes reduced macrophage infiltration and pro-inflammatory response. Conclusions: The NaA@SC/MAN-LPs demonstrated effective dual targeting effects on hepatocytes and Kupffer cells, significantly improving the pathogenesis of NAFLD, compared to free NaA. This study provides a new strategy for developing effective and safe oral drugs for NAFLD.
Collapse
Affiliation(s)
- Yichao Hou
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
| | - Xilong Gao
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
| | - Jiahui Gong
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (J.G.); (X.D.); (Z.Z.); (H.Z.)
| | - Xinrui Dong
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (J.G.); (X.D.); (Z.Z.); (H.Z.)
| | - Yanling Hao
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
| | - Zhengyuan Zhai
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (J.G.); (X.D.); (Z.Z.); (H.Z.)
| | - Hao Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (J.G.); (X.D.); (Z.Z.); (H.Z.)
| | - Ming Zhang
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China;
| | - Rong Liu
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
| | - Ran Wang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
- Research Center for Probiotics, China Agricultural University, Beijing 101299, China
| | - Liang Zhao
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (J.G.); (X.D.); (Z.Z.); (H.Z.)
- Research Center for Probiotics, China Agricultural University, Beijing 101299, China
| |
Collapse
|
5
|
Igudesman D, Yu G, Dutta T, Carnero EA, Krajmalnik-Brown R, Smith SR, Corbin KD. Global metabolite profiling in feces, serum, and urine yields insights into energy balance phenotypes induced by diet-driven microbiome remodeling. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.05.25321733. [PMID: 39974023 PMCID: PMC11838622 DOI: 10.1101/2025.02.05.25321733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Background Preclinical literature and behavioral human data suggest that diet profoundly impacts the human gut microbiome and energy absorption-a key determinant of energy balance. To determine whether these associations are causal, domiciled controlled feeding studies with precise measurements of dietary intake and energy balance are needed. Metabolomics-a functional readout of microbiome modulation-can help identify putative mechanisms mediating these effects. We previously demonstrated that a high-fiber, minimally processed Microbiome Enhancer Diet (MBD) fed at energy balance decreased energy absorption and increased microbial biomass relative to a calorie-matched fiber-poor, highly processed Western Diet (WD). Objective To identify metabolic signatures distinguishing MBD from WD feeding and potential metabolomic mechanisms mediating the MBD-induced negative energy balance. Methods We deployed global metabolomics in feces, serum, and urine using samples collected at the end of a randomized crossover controlled feeding trial delivering 22 days of an MBD and a WD to 17 persons without obesity. Samples were collected while participants were domiciled on a metabolic ward and analyzed using Ultrahigh Performance Liquid Chromatography-Tandem Mass Spectroscopy. Linear mixed effects models tested metabolite changes by diet. Weighted gene network correlation analysis identified metabolite modules correlated with energy balance phenotypes. Results Numerous metabolites consistently altered in the feces, fasting serum, and/or urine may serve as putative dietary biomarkers of MBD feeding. Fecal diet-microbiota co-metabolites decreased by an MBD correlated with reduced energy absorption and increased microbial biomass. An MBD shifted the urinary metabolome from sugar degradation to ketogenesis-evidence of negative energy balance. Conclusions Precisely controlled diets disparate in microbiota-accessible substrates led to distinct metabolomic signatures in feces, fasting serum, and/or urine. These diet-microbiota co-metabolites may be biomarkers of a "fed" (MBD) or "starved" (WD) gut microbiota associated with energy balance. These findings lay the foundation for unveiling causal pathways linking diet-microbiota co-metabolism to energy absorption.
Collapse
|
6
|
Chong S, Lin M, Chong D, Jensen S, Lau NS. A systematic review on gut microbiota in type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2025; 15:1486793. [PMID: 39897957 PMCID: PMC11782031 DOI: 10.3389/fendo.2024.1486793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/18/2024] [Indexed: 02/04/2025] Open
Abstract
Aims/hypothesis The gut microbiota play crucial roles in the digestion and degradation of nutrients, synthesis of biological agents, development of the immune system, and maintenance of gastrointestinal integrity. Gut dysbiosis is thought to be associated with type 2 diabetes mellitus (T2DM), one of the world's fastest growing diseases. The aim of this systematic review is to identify differences in the composition and diversity of the gut microbiota in individuals with T2DM. Methods A systematic search was conducted to identify studies reporting on the difference in gut microbiota composition between individuals with T2DM and healthy controls. Relevant studies were evaluated, and their characteristics and results were extracted using a standardized data extraction form. The studies were assessed for risk of bias and their findings were reported narratively. Results 58 observational studies published between 2010 and 2024 were included. Beta diversity was commonly reported to be different between individuals with T2DM and healthy individuals. Genera Lactobacillus, Escherichia-Shigella, Enterococcus, Subdoligranulum and Fusobacteria were found to be positively associated; while Akkermansia, Bifidobacterium, Bacteroides, Roseburia, Faecalibacteirum and Prevotella were found to be negatively associated with T2DM. Conclusions This systematic review demonstrates a strong association between T2DM and gut dysbiosis, as evidenced by differential microbial abundances and altered diversity indices. Among these taxa, Escherichia-Shigella is consistently associated with T2DM, whereas Faecalibacterium prausnitzii appears to offer a protective effect against T2DM. However, the heterogeneity and observational nature of these studies preclude the establishment of causative relationships. Future research should incorporate age, diet and medication-matched controls, and include functional analysis of these gut microbes. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42023459937.
Collapse
Affiliation(s)
- Serena Chong
- South West Sydney Limb Preservation and Wound Research, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
- South West Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Mike Lin
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
- Garvan Institute of Research, Sydney, NSW, Australia
| | - Deborah Chong
- Animal Health Laboratory, Department of Natural Resources and Environment Tasmania, Tasmania, TAS, Australia
| | - Slade Jensen
- South West Sydney Limb Preservation and Wound Research, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
- Infectious Disease and Microbiology, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
- School of Medicine Antibiotic Resistance and Mobile Elements Groups, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
| | - Namson S. Lau
- South West Sydney Limb Preservation and Wound Research, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
- South West Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Liverpool Diabetes Collaboration, Ingham Institute of Applied Medical Research, Sydney, NSW, Australia
| |
Collapse
|
7
|
Fang H, Rodrigues e-Lacerda R, Barra NG, Kukje Zada D, Robin N, Mehra A, Schertzer JD. Postbiotic Impact on Host Metabolism and Immunity Provides Therapeutic Potential in Metabolic Disease. Endocr Rev 2025; 46:60-79. [PMID: 39235984 PMCID: PMC11720174 DOI: 10.1210/endrev/bnae025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/18/2024] [Accepted: 09/04/2024] [Indexed: 09/07/2024]
Abstract
The gut microbiota influences aspects of metabolic disease, including tissue inflammation, adiposity, blood glucose, insulin, and endocrine control of metabolism. Prebiotics or probiotics are often sought to combat metabolic disease. However, prebiotics lack specificity and can have deleterious bacterial community effects. Probiotics require live bacteria to find a colonization niche sufficient to influence host immunity or metabolism. Postbiotics encompass bacterial-derived components and molecules, which are well-positioned to alter host immunometabolism without relying on colonization efficiency or causing widespread effects on the existing microbiota. Here, we summarize the potential for beneficial and detrimental effects of specific postbiotics related to metabolic disease and the underlying mechanisms of action. Bacterial cell wall components, such as lipopolysaccharides, muropeptides, lipoteichoic acids and flagellin, have context-dependent effects on host metabolism by engaging specific immune responses. Specific types of postbiotics within broad classes of compounds, such as lipopolysaccharides and muropeptides, can have opposing effects on endocrine control of host metabolism, where certain postbiotics are insulin sensitizers and others promote insulin resistance. Bacterial metabolites, such as short-chain fatty acids, bile acids, lactate, glycerol, succinate, ethanolamine, and ethanol, can be substrates for host metabolism. Postbiotics can fuel host metabolic pathways directly or influence endocrine control of metabolism through immunomodulation or mimicking host-derived hormones. The interaction of postbiotics in the host-microbe relationship should be considered during metabolic inflammation and metabolic disease.
Collapse
Affiliation(s)
- Han Fang
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Rodrigo Rodrigues e-Lacerda
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Nicole G Barra
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Dana Kukje Zada
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Nazli Robin
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Alina Mehra
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| |
Collapse
|
8
|
Rowghani K, Patel B, Martinez-Guryn K. Dietary impact on the gut microbiome and epigenome and regulation of gut inflammation. NUTRITION IN THE CONTROL OF INFLAMMATION 2025:369-398. [DOI: 10.1016/b978-0-443-18979-1.00014-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Green CG, Ong MLY, Rowland SN, Bongiovanni T, James LJ, Clifford T, Bailey SJ, Heaney LM. Investigating serum concentration profiles of orally ingested short-chain fatty acid supplements. Food Funct 2024; 15:11525-11536. [PMID: 39498577 DOI: 10.1039/d4fo04028g] [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: 11/26/2024]
Abstract
Acetate, propionate, and butyrate are naturally-occurring short-chain fatty acids (SCFAs) derived from bacterial metabolism of dietary fibre and have been associated with numerous positive health outcomes. All three acids have been shown to offer unique physiological and metabolic effects and, therefore, could be targeted for co-ingestion as part of a nutritional/medicinal plan. However, a better understanding of the outcomes of supplementing in combination on circulating concentration profiles is necessary to confirm uptake efficacy. This study sought to investigate the acute circulating concentration profiles of acetate, propionate, and butyrate following oral supplementation. Three experimental trials were conducted including investigations to understand the impact of capsule coating on circulating concentration profiles, the effect of supplementation dose on uptake kinetics, and the outcome of a short, repeated, supplementation routine on circulating levels. Serum samples were analysed for SCFA content using a quantitative GC-MS assay. It was observed that an acid-resistant coated capsule caused a delayed and blunted blood concentration response, with the non-acid resistant trial displaying earlier and more intense peak serum concentrations. For dose comparison investigations, all SCFAs peaked within 60 min and returned to baseline concentrations by 120 min post-supplementation. A graded dose relationship was present for propionate and butyrate when considering the total circulating exposure across a 240 min monitoring period. In addition, a one-week, twice-daily, repeated supplementation protocol resulted in no changes in basal serum SCFA concentrations. Overall, these data indicate that acetate, propionate, and butyrate display relatively similar circulating concentration profiles following oral co-ingestion, adding knowledge to help inform supplementation strategies for future outcomes where acute elevation of circulating SCFAs is desired.
Collapse
Affiliation(s)
- Christopher G Green
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| | - Marilyn L Y Ong
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
- Exercise and Sports Science Programme, School of Health Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Samantha N Rowland
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| | - Tindaro Bongiovanni
- Player Health & Performance Department, Palermo Football Club, Palermo, Italy
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Lewis J James
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| | - Tom Clifford
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| | - Stephen J Bailey
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| | - Liam M Heaney
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| |
Collapse
|
10
|
Rhimi S, Jablaoui A, Hernandez J, Mariaule V, Akermi N, Méric T, Mkaouar H, Wysocka M, Lesner A, Borgi MA, Maguin E, Rhimi M. Industrial diet intervention modulates the interplay between gut microbiota and host in semi-stray dogs. Anim Microbiome 2024; 6:69. [PMID: 39574203 PMCID: PMC11580502 DOI: 10.1186/s42523-024-00357-w] [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: 03/28/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND The gut microbiota and derived metabolites play a key role in regulating host physiology. Diet is identified as a key regulatory factor of the microbiota composition and, potentially, of subsequent functionalities. Demonstrating the role of diet may be complex as most human studies are cross-sectional and dietary intervention is often accompanied by hygienic changes. The objective of the present study was to investigate the impact of an industrial diet on the modulation of the microbiota and targeted functionalities using a canine "natural" model. RESULTS We carried out a controlled dietary trial in a cohort of Tunisian semi-stray dogs. We made a transition from a natural diet to an industrial kibble diet and monitored the composition of the fecal microbiota, the concentration of short-chain fatty acids (SCFA) and bile acids (BAs), and protease activities. We demonstrated that dietary change significantly decreased fecal primary bile acids levels and protease activities. Interestingly, correlation analyses demonstrated that variation of specific microbial genera were associated with modulated physiological parameters. CONCLUSIONS Our study reveals that an industrial diet induces beneficial changes in microbial composition and functions characterised by increased diversity, synthesis of SCFA and secondary bile acids production, stressing the key role of the diet-microbiota-dog crosstalk.
Collapse
Affiliation(s)
- Soufien Rhimi
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, Institut National de Recherche Pour l'Agriculture, l'Alimentation et l'Environnement, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
- Laboratory of Biotechnology and Biomonitoring of the Environment and Oasis Ecosystems (LBBEEO), Faculty of Sciences of Gafsa, University of Gafsa, Zarroug, 2112, Gafsa, Tunisia
| | - Amin Jablaoui
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, Institut National de Recherche Pour l'Agriculture, l'Alimentation et l'Environnement, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Juan Hernandez
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, Institut National de Recherche Pour l'Agriculture, l'Alimentation et l'Environnement, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
- Oniris VetAgroBio Nantes, Department of Clinical Sciences, Nantes-Atlantic College of Veterinary Medicine and Food Sciences, 44300, Nantes, France
| | - Vincent Mariaule
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, Institut National de Recherche Pour l'Agriculture, l'Alimentation et l'Environnement, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Nizar Akermi
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, Institut National de Recherche Pour l'Agriculture, l'Alimentation et l'Environnement, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Tristan Méric
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, Institut National de Recherche Pour l'Agriculture, l'Alimentation et l'Environnement, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
- Oniris VetAgroBio Nantes, Department of Clinical Sciences, Nantes-Atlantic College of Veterinary Medicine and Food Sciences, 44300, Nantes, France
| | - Héla Mkaouar
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, Institut National de Recherche Pour l'Agriculture, l'Alimentation et l'Environnement, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Magdalena Wysocka
- Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80308, Gdańsk, Poland
| | - Adam Lesner
- Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80308, Gdańsk, Poland
| | - Mohamed Ali Borgi
- Laboratory of Biotechnology and Biomonitoring of the Environment and Oasis Ecosystems (LBBEEO), Faculty of Sciences of Gafsa, University of Gafsa, Zarroug, 2112, Gafsa, Tunisia
| | - Emmanuelle Maguin
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, Institut National de Recherche Pour l'Agriculture, l'Alimentation et l'Environnement, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France.
| | - Moez Rhimi
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, Institut National de Recherche Pour l'Agriculture, l'Alimentation et l'Environnement, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France.
| |
Collapse
|
11
|
Eslami M, Pakmehr A, Pourghazi F, Kami A, Ejtahed HS, Mohajeri-Tehrani M, Hasani-Ranjbar S, Larijani B. The anti-obesity effects of postbiotics: A systematic review of pre-clinical and clinical studies. Clin Nutr ESPEN 2024; 64:370-389. [PMID: 39461594 DOI: 10.1016/j.clnesp.2024.10.153] [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: 06/25/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND The growing prevalence of obesity has become a major concern worldwide, therefore a great number of studies are conducted every day in the field of obesity. Since postbiotics are a newly introduced term, there is not much systematic evidence about their function and impact on obesity. We designed this study to systematically review the effect of different types of postbiotics on obesity. METHODS A systematic search was conducted using PubMed, SCOPUS, and Web of Science databases up to August 2023. Both human and animal interventional studies that investigated the effects of any type of postbiotic on obesity and obesity-related factors were eligible. Screening, data extraction, and quality assessment were conducted independently by two researchers. The quality of the studies was appraised using Cochrane and Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE's) risk of bias tool. RESULTS Of the 19373 retrieved studies, finally, 49 studies were included (9 human studies and 40 animal studies). Short-chain fatty acids and heat-killed (inactivated) bacteria were the most used postbiotics. In human clinical trials, inactivated Lactobacillus amylovorus (CP1563), Bifidobacterium animalis subsp. lactis (CECT 8145) and Pediococcus pentosaceus (LP28) were administered orally as postbiotics which improved body composition and anthropometric indices. Animal studies evaluated other types of postbiotics including muramyl dipeptide, cell-free extracts, urolithin A&B, extracellular Vesicles, exopolysaccharides, and surface Layer Proteins, supporting the anti-obesity effects of postbiotics. CONCLUSION Postbiotics seem to be a safe intervention and the results were in favor of a reduction in adipogenesis as well as an increase in energy expenditure. Further high-quality studies are required in this relatively new topic.
Collapse
Affiliation(s)
- Maysa Eslami
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Azin Pakmehr
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzad Pourghazi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Atefe Kami
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanieh-Sadat Ejtahed
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammadreza Mohajeri-Tehrani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirin Hasani-Ranjbar
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Kirtipal N, Seo Y, Son J, Lee S. Systems Biology of Human Microbiome for the Prediction of Personal Glycaemic Response. Diabetes Metab J 2024; 48:821-836. [PMID: 39313228 PMCID: PMC11449821 DOI: 10.4093/dmj.2024.0382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
The human gut microbiota is increasingly recognized as a pivotal factor in diabetes management, playing a significant role in the body's response to treatment. However, it is important to understand that long-term usage of medicines like metformin and other diabetic treatments can result in problems, gastrointestinal discomfort, and dysbiosis of the gut flora. Advanced sequencing technologies have improved our understanding of the gut microbiome's role in diabetes, uncovering complex interactions between microbial composition and metabolic health. We explore how the gut microbiota affects glucose metabolism and insulin sensitivity by examining a variety of -omics data, including genomics, transcriptomics, epigenomics, proteomics, metabolomics, and metagenomics. Machine learning algorithms and genome-scale modeling are now being applied to find microbiological biomarkers associated with diabetes risk, predicted disease progression, and guide customized therapy. This study holds promise for specialized diabetic therapy. Despite significant advances, some concerns remain unanswered, including understanding the complex relationship between diabetes etiology and gut microbiota, as well as developing user-friendly technological innovations. This mini-review explores the relationship between multiomics, precision medicine, and machine learning to improve our understanding of the gut microbiome's function in diabetes. In the era of precision medicine, the ultimate goal is to improve patient outcomes through personalized treatments.
Collapse
Affiliation(s)
- Nikhil Kirtipal
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Youngchang Seo
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Jangwon Son
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Bucheon, Korea
| | - Sunjae Lee
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| |
Collapse
|
13
|
Byndloss M, Devkota S, Duca F, Niess JH, Nieuwdorp M, Orho-Melander M, Sanz Y, Tremaroli V, Zhao L. The gut microbiota and diabetes: research, translation, and clinical applications - 2023 Diabetes, Diabetes Care, and Diabetologia Expert Forum. Diabetologia 2024; 67:1760-1782. [PMID: 38910152 PMCID: PMC11410996 DOI: 10.1007/s00125-024-06198-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/23/2024] [Indexed: 06/25/2024]
Abstract
This article summarises the state of the science on the role of the gut microbiota (GM) in diabetes from a recent international expert forum organised by Diabetes, Diabetes Care, and Diabetologia, which was held at the European Association for the Study of Diabetes 2023 Annual Meeting in Hamburg, Germany. Forum participants included clinicians and basic scientists who are leading investigators in the field of the intestinal microbiome and metabolism. Their conclusions were as follows: (1) the GM may be involved in the pathophysiology of type 2 diabetes, as microbially produced metabolites associate both positively and negatively with the disease, and mechanistic links of GM functions (e.g. genes for butyrate production) with glucose metabolism have recently emerged through the use of Mendelian randomisation in humans; (2) the highly individualised nature of the GM poses a major research obstacle, and large cohorts and a deep-sequencing metagenomic approach are required for robust assessments of associations and causation; (3) because single time point sampling misses intraindividual GM dynamics, future studies with repeated measures within individuals are needed; and (4) much future research will be required to determine the applicability of this expanding knowledge to diabetes diagnosis and treatment, and novel technologies and improved computational tools will be important to achieve this goal.
Collapse
Affiliation(s)
- Mariana Byndloss
- Vanderbilt University Medical Center, Nashville, TN, USA
- Howard Hughes Medical Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Suzanne Devkota
- Cedars-Sinai Medical Center, Human Microbiome Research Institute, Los Angeles, CA, USA
| | | | - Jan Hendrik Niess
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Department of Gastroenterology and Hepatology, University Digestive Healthcare Center, Clarunis, Basel, Switzerland
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
- Amsterdam Diabeter Center, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Marju Orho-Melander
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Yolanda Sanz
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain.
| | - Valentina Tremaroli
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Liping Zhao
- Department of Biochemistry and Microbiology, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ, USA
| |
Collapse
|
14
|
Larik GNF, Canfora EE, van Schothorst EM, Blaak EE. Intestinal gases as a non-invasive measurement of microbial fermentation and host health. Cell Host Microbe 2024; 32:1225-1229. [PMID: 39146794 DOI: 10.1016/j.chom.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 08/17/2024]
Abstract
Microbial fermentation and associated products provide insights into the gut microbiota-host relationship. Here, we propose using improved technologies that allow non-invasive, real-time measurements of intestinal gases as a metric for microbial fermentation. This approach has the potential to provide a basis for personalized interventions that improve host metabolic health.
Collapse
Affiliation(s)
- Gillian N F Larik
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Emanuel E Canfora
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Evert M van Schothorst
- Human and Animal Physiology, Wageningen University & Research, Wageningen, the Netherlands
| | - Ellen E Blaak
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, the Netherlands.
| |
Collapse
|
15
|
Ren Y, Huang P, Zhang L, Tang YF, Luo SL, She Z, Peng H, Chen YQ, Luo JW, Duan WX, Liu LJ, Liu LQ. Dual Regulation Mechanism of Obesity: DNA Methylation and Intestinal Flora. Biomedicines 2024; 12:1633. [PMID: 39200098 PMCID: PMC11351752 DOI: 10.3390/biomedicines12081633] [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: 06/12/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 09/01/2024] Open
Abstract
Obesity is a multifactorial chronic inflammatory metabolic disorder, with pathogenesis influenced by genetic and non-genetic factors such as environment and diet. Intestinal microbes and their metabolites play significant roles in the occurrence and development of obesity by regulating energy metabolism, inducing chronic inflammation, and impacting intestinal hormone secretion. Epigenetics, which involves the regulation of host gene expression without changing the nucleotide sequence, provides an exact direction for us to understand how the environment, lifestyle factors, and other risk factors contribute to obesity. DNA methylation, as the most common epigenetic modification, is involved in the pathogenesis of various metabolic diseases. The epigenetic modification of the host is induced or regulated by the intestinal microbiota and their metabolites, linking the dynamic interaction between the microbiota and the host genome. In this review, we examined recent advancements in research, focusing on the involvement of intestinal microbiota and DNA methylation in the etiology and progression of obesity, as well as potential interactions between the two factors, providing novel perspectives and avenues for further elucidating the pathogenesis, prevention, and treatment of obesity.
Collapse
Affiliation(s)
- Yi Ren
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
- Department of Pediatrics, Haikou Hospital of the Maternal and Child Health, Haikou 570100, China
- Department of Children’s Healthcare, Hainan Modern Women and Children’s Medical, Haikou 570100, China
| | - Peng Huang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Lu Zhang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yu-Fen Tang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Sen-Lin Luo
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Zhou She
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Hong Peng
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yu-Qiong Chen
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Jin-Wen Luo
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Wang-Xin Duan
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Ling-Juan Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Li-Qun Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| |
Collapse
|
16
|
Vliex LMM, Penders J, Nauta A, Zoetendal EG, Blaak EE. The individual response to antibiotics and diet - insights into gut microbial resilience and host metabolism. Nat Rev Endocrinol 2024; 20:387-398. [PMID: 38486011 DOI: 10.1038/s41574-024-00966-0] [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] [Accepted: 02/20/2024] [Indexed: 06/16/2024]
Abstract
Antibiotic use disrupts microbial composition and activity in humans, but whether this disruption in turn affects host metabolic health is unclear. Cohort studies show associations between antibiotic use and an increased risk of developing obesity and type 2 diabetes mellitus. Here, we review available clinical trials and show the disruptive effect of antibiotic use on the gut microbiome in humans, as well as its impact on bile acid metabolism and microbial metabolites such as short-chain fatty acids. Placebo-controlled human studies do not show a consistent effect of antibiotic use on body weight and insulin sensitivity at a population level, but rather an individual-specific or subgroup-specific response. This response to antibiotic use is affected by the resistance and resilience of the gut microbiome, factors that determine the extent of disruption and the speed of recovery afterwards. Nutritional strategies to improve the composition and functionality of the gut microbiome, as well as its recovery after antibiotic use (for instance, with prebiotics), require a personalized approach to increase their efficacy. Improved insights into key factors that influence the individual-specific response to antibiotics and dietary intervention may lead to better efficacy in reversing or preventing antibiotic-induced microbial dysbiosis as well as strategies for preventing cardiometabolic diseases.
Collapse
Affiliation(s)
- Lars M M Vliex
- Department of Human Biology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Arjen Nauta
- FrieslandCampina, Amersfoort, The Netherlands
| | - Erwin G Zoetendal
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands.
| |
Collapse
|
17
|
Boven L, Akkerman R, de Vos P. Sustainable diets with plant-based proteins require considerations for prevention of proteolytic fermentation. Crit Rev Food Sci Nutr 2024; 65:2829-2839. [PMID: 38950600 DOI: 10.1080/10408398.2024.2352523] [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] [Indexed: 07/03/2024]
Abstract
The human diet requires a more plant-based approach due to the exhaustive effects animal-based foods have on the environment. However, plant-based proteins generally miss a few or have a lower variety in essential amino acids and are more difficult to digest. Subsequently they might be prone to fermentation by the microbiome in the proximal colon. Proteolytic fermentation can induce microbial-metabolites with beneficial and negative health effects. We review current insight into how balances in saccharolytic and proteolytic fermentation can be maintained when the diet consists predominantly of plant-based proteins. Some proteolytic fermentation metabolites may negatively impact balances in gut microbiota composition in the large intestine and influence immunity. However, proteolytic fermentation can potentially be prevented in the proximal colon toward more saccharolytic fermentation through the addition of non-digestible carbohydrates in the diet. Knowledge on this combination of plant-based proteins and non-digestible carbohydrates on colonic- and general health is limited. Current data suggest that transitioning toward a more plant-based protein diet should be accompanied with a consumption of increased quantities and more complex structures of carbohydrates or by application of technological strategies to enhances digestibility. This can reduce or prevent proteolytic fermentation which might consequently improve human health.
Collapse
Affiliation(s)
- Lidwien Boven
- Immunoendocrinology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Renate Akkerman
- Immunoendocrinology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Paul de Vos
- Immunoendocrinology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
18
|
Lou X, Li P, Luo X, Lei Z, Liu X, Liu Y, Gao L, Xu W, Liu X. Dietary patterns interfere with gut microbiota to combat obesity. Front Nutr 2024; 11:1387394. [PMID: 38953044 PMCID: PMC11215203 DOI: 10.3389/fnut.2024.1387394] [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: 02/17/2024] [Accepted: 06/07/2024] [Indexed: 07/03/2024] Open
Abstract
Obesity and obesity-related metabolic disorders are global epidemics that occur when there is chronic energy intake exceeding energy expenditure. Growing evidence suggests that healthy dietary patterns not only decrease the risk of obesity but also influence the composition and function of the gut microbiota. Numerous studies manifest that the development of obesity is associated with gut microbiota. One promising supplementation strategy is modulating gut microbiota composition by dietary patterns to combat obesity. In this review, we discuss the changes of gut microbiota in obesity and obesity-related metabolic disorders, with a particular emphasis on the impact of dietary components on gut microbiota and how common food patterns can intervene in gut microbiota to prevent obesity. While there is promise in intervening with the gut microbiota to combat obesity through the regulation of dietary patterns, numerous key questions remain unanswered. In this review, we critically review the associations between dietary patterns, gut microbes, and obesity, aiming to contribute to the further development and application of dietary patterns against obesity in humans.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xiaomeng Liu
- Nutrition and Food Hygiene Laboratory, School of Public Health, Xinxiang Medical College, Xinxiang, China
| |
Collapse
|
19
|
Hijová E. Postbiotics as Metabolites and Their Biotherapeutic Potential. Int J Mol Sci 2024; 25:5441. [PMID: 38791478 PMCID: PMC11121590 DOI: 10.3390/ijms25105441] [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: 04/02/2024] [Revised: 05/06/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
This review highlights the role of postbiotics, which may provide an underappreciated avenue doe promising therapeutic alternatives. The discovery of natural compounds obtained from microorganisms needs to be investigated in the future in terms of their effects on various metabolic disorders and molecular pathways, as well as modulation of the immune system and intestinal microbiota in children and adults. However, further studies and efforts are needed to evaluate and describe new postbiotics. This review provides available knowledge that may assist future research in identifying new postbiotics and uncovering additional mechanisms to combat metabolic diseases.
Collapse
Affiliation(s)
- Emília Hijová
- Center of Clinical and Preclinical Research MEDIPARK, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia
| |
Collapse
|
20
|
Deehan EC, Mocanu V, Madsen KL. Effects of dietary fibre on metabolic health and obesity. Nat Rev Gastroenterol Hepatol 2024; 21:301-318. [PMID: 38326443 DOI: 10.1038/s41575-023-00891-z] [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] [Accepted: 12/18/2023] [Indexed: 02/09/2024]
Abstract
Obesity and metabolic syndrome represent a growing epidemic worldwide. Body weight is regulated through complex interactions between hormonal, neural and metabolic pathways and is influenced by numerous environmental factors. Imbalances between energy intake and expenditure can occur due to several factors, including alterations in eating behaviours, abnormal satiation and satiety, and low energy expenditure. The gut microbiota profoundly affects all aspects of energy homeostasis through diverse mechanisms involving effects on mucosal and systemic immune, hormonal and neural systems. The benefits of dietary fibre on metabolism and obesity have been demonstrated through mechanistic studies and clinical trials, but many questions remain as to how different fibres are best utilized in managing obesity. In this Review, we discuss the physiochemical properties of different fibres, current findings on how fibre and the gut microbiota interact to regulate body weight homeostasis, and knowledge gaps related to using dietary fibres as a complementary strategy. Precision medicine approaches that utilize baseline microbiota and clinical characteristics to predict individual responses to fibre supplementation represent a new paradigm with great potential to enhance weight management efficacy, but many challenges remain before these approaches can be fully implemented.
Collapse
Affiliation(s)
- Edward C Deehan
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, USA
- Nebraska Food for Health Center, Lincoln, NE, USA
| | - Valentin Mocanu
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Karen L Madsen
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
21
|
Facchin S, Bertin L, Bonazzi E, Lorenzon G, De Barba C, Barberio B, Zingone F, Maniero D, Scarpa M, Ruffolo C, Angriman I, Savarino EV. Short-Chain Fatty Acids and Human Health: From Metabolic Pathways to Current Therapeutic Implications. Life (Basel) 2024; 14:559. [PMID: 38792581 PMCID: PMC11122327 DOI: 10.3390/life14050559] [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: 03/25/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
The gastrointestinal tract is home to trillions of diverse microorganisms collectively known as the gut microbiota, which play a pivotal role in breaking down undigested foods, such as dietary fibers. Through the fermentation of these food components, short-chain fatty acids (SCFAs) such as acetate, propionate, and butyrate are produced, offering numerous health benefits to the host. The production and absorption of these SCFAs occur through various mechanisms within the human intestine, contingent upon the types of dietary fibers reaching the gut and the specific microorganisms engaged in fermentation. Medical literature extensively documents the supplementation of SCFAs, particularly butyrate, in the treatment of gastrointestinal, metabolic, cardiovascular, and gut-brain-related disorders. This review seeks to provide an overview of the dynamics involved in the production and absorption of acetate, propionate, and butyrate within the human gut. Additionally, it will focus on the pivotal roles these SCFAs play in promoting gastrointestinal and metabolic health, as well as their current therapeutic implications.
Collapse
Affiliation(s)
- Sonia Facchin
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Luisa Bertin
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Erica Bonazzi
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Greta Lorenzon
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Caterina De Barba
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Brigida Barberio
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Fabiana Zingone
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Daria Maniero
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Marco Scarpa
- General Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35138 Padua, Italy (C.R.); (I.A.)
| | - Cesare Ruffolo
- General Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35138 Padua, Italy (C.R.); (I.A.)
| | - Imerio Angriman
- General Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35138 Padua, Italy (C.R.); (I.A.)
| | - Edoardo Vincenzo Savarino
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| |
Collapse
|
22
|
Mishra N, Garg A, Ashique S, Bhatt S. Potential of postbiotics for the treatment of metabolic disorders. Drug Discov Today 2024; 29:103921. [PMID: 38382867 DOI: 10.1016/j.drudis.2024.103921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/07/2024] [Accepted: 02/15/2024] [Indexed: 02/23/2024]
Abstract
Postbiotics, the next generation of probiotics, are extracts that are free of living and nonviable bacteria and show strong modulatory effects on the gut flora. Examples include vitamin B12, vitamin K, folate, lipopolysaccharides, enzymes, and short-chain fatty acids (SCFAs), representing a subset of essential nutrients commonly found in the human diet. Postbiotics have been observed to demonstrate antiobesity and antidiabetic effects through a variety of mechanisms. These pathways primarily involve an elevation in energy expenditure, a decrease in the formation and differentiation of adipocytes and food intake, modification of lipid and carbohydrate absorption and metabolism, and regulation of gut dysbiosis. Based on these above effects and mechanisms, the use of postbiotics can be considered as potential strategy for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Neeraj Mishra
- Amity Institute of Pharmacy, Amity University Madhya Pradesh, Gwalior 474005, India
| | - Ashish Garg
- Department of Pharmaceutics, Guru RamdasKhalsa Institute of Science and Technology (Pharmacy), Jabalpur 483001, Madhya Pradesh, India
| | - Sumel Ashique
- Department of Pharmaceutical Sciences, Bengal College of Pharmaceutical Sciences & Research, Durgapur 713212, West Bengal, India
| | - Shvetank Bhatt
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, Dr. Vishwanath Karad MIT World Peace University, Pune 411038, Maharashtra, India.
| |
Collapse
|
23
|
Williams LM, Cao S. Harnessing and delivering microbial metabolites as therapeutics via advanced pharmaceutical approaches. Pharmacol Ther 2024; 256:108605. [PMID: 38367866 PMCID: PMC10985132 DOI: 10.1016/j.pharmthera.2024.108605] [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: 10/31/2023] [Revised: 01/05/2024] [Accepted: 02/08/2024] [Indexed: 02/19/2024]
Abstract
Microbial metabolites have emerged as key players in the interplay between diet, the gut microbiome, and host health. Two major classes, short-chain fatty acids (SCFAs) and tryptophan (Trp) metabolites, are recognized to regulate inflammatory, immune, and metabolic responses within the host. Given that many human diseases are associated with dysbiosis of the gut microbiome and consequent reductions in microbial metabolite production, the administration of these metabolites represents a direct, multi-targeted treatment. While a multitude of preclinical studies showcase the therapeutic potential of both SCFAs and Trp metabolites, they often rely on high doses and frequent dosing regimens to achieve systemic effects, thereby constraining their clinical applicability. To address these limitations, a variety of pharmaceutical formulations approaches that enable targeted, delayed, and/or sustained microbial metabolite delivery have been developed. These approaches, including enteric encapsulations, esterification to dietary fiber, prodrugs, and nanoformulations, pave the way for the next generation of microbial metabolite-based therapeutics. In this review, we first provide an overview of the roles of microbial metabolites in maintaining host homeostasis and outline how compromised metabolite production contributes to the pathogenesis of inflammatory, metabolic, autoimmune, allergic, infectious, and cancerous diseases. Additionally, we explore the therapeutic potential of metabolites in these disease contexts. Then, we provide a comprehensive and up-to-date review of the pharmaceutical strategies that have been employed to enhance the therapeutic efficacy of microbial metabolites, with a focus on SCFAs and Trp metabolites.
Collapse
Affiliation(s)
- Lindsey M Williams
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, United States
| | - Shijie Cao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, United States.
| |
Collapse
|
24
|
Apalowo OE, Adegoye GA, Obuotor TM. Microbial-Based Bioactive Compounds to Alleviate Inflammation in Obesity. Curr Issues Mol Biol 2024; 46:1810-1831. [PMID: 38534735 DOI: 10.3390/cimb46030119] [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: 02/07/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/28/2024] Open
Abstract
The increased prevalence of obesity with several other metabolic disorders, including diabetes and non-alcoholic fatty liver disease, has reached global pandemic proportions. Lifestyle changes may result in a persistent positive energy balance, hastening the onset of these age-related disorders and consequently leading to a diminished lifespan. Although suggestions have been raised on the possible link between obesity and the gut microbiota, progress has been hampered due to the extensive diversity and complexities of the gut microbiota. Being recognized as a potential biomarker owing to its pivotal role in metabolic activities, the dysregulation of the gut microbiota can give rise to a persistent low-grade inflammatory state associated with chronic diseases during aging. This chronic inflammatory state, also known as inflammaging, induced by the chronic activation of the innate immune system via the macrophage, is controlled by the gut microbiota, which links nutrition, metabolism, and the innate immune response. Here, we present the functional roles of prebiotics, probiotics, synbiotics, and postbiotics as bioactive compounds by underscoring their putative contributions to (1) the reduction in gut hyperpermeability due to lipopolysaccharide (LPS) inactivation, (2) increased intestinal barrier function as a consequence of the upregulation of tight junction proteins, and (3) inhibition of proinflammatory pathways, overall leading to the alleviation of chronic inflammation in the management of obesity.
Collapse
Affiliation(s)
- Oladayo Emmanuel Apalowo
- Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Starkville, MS 39762, USA
| | - Grace Adeola Adegoye
- Department of Nutrition and Health Science, Ball State University, Muncie, IN 47306, USA
| | | |
Collapse
|
25
|
Nayman EI, Schwartz BA, Polmann M, Gumabong AC, Nieuwdorp M, Cickovski T, Mathee K. Differences in gut microbiota between Dutch and South-Asian Surinamese: potential implications for type 2 diabetes mellitus. Sci Rep 2024; 14:4585. [PMID: 38403716 PMCID: PMC10894869 DOI: 10.1038/s41598-024-54769-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 02/16/2024] [Indexed: 02/27/2024] Open
Abstract
Gut microbiota, or the collection of diverse microorganisms in a specific ecological niche, are known to significantly impact human health. Decreased gut microbiota production of short-chain fatty acids (SCFAs) has been implicated in type 2 diabetes mellitus (T2DM) disease progression. Most microbiome studies focus on ethnic majorities. This study aims to understand how the microbiome differs between an ethnic majority (the Dutch) and minority (the South-Asian Surinamese (SAS)) group with a lower and higher prevalence of T2DM, respectively. Microbiome data from the Healthy Life in an Urban Setting (HELIUS) cohort were used. Two age- and gender-matched groups were compared: the Dutch (n = 41) and SAS (n = 43). Microbial community compositions were generated via DADA2. Metrics of microbial diversity and similarity between groups were computed. Biomarker analyses were performed to determine discriminating taxa. Bacterial co-occurrence networks were constructed to examine ecological patterns. A tight microbiota cluster was observed in the Dutch women, which overlapped with some of the SAS microbiota. The Dutch gut contained a more interconnected microbial ecology, whereas the SAS network was dispersed, i.e., contained fewer inter-taxonomic correlational relationships. Bacteroides caccae, Butyricicoccus, Alistipes putredinis, Coprococcus comes, Odoribacter splanchnicus, and Lachnospira were enriched in the Dutch gut. Haemophilus, Bifidobacterium, and Anaerostipes hadrus discriminated the SAS gut. All but Lachnospira and certain strains of Haemophilus are known to produce SCFAs. The Dutch gut microbiome was distinguished from the SAS by diverse, differentially abundant SCFA-producing taxa with significant cooperation. The dynamic ecology observed in the Dutch was not detected in the SAS. Among several potential gut microbial biomarkers, Haemophilus parainfluenzae likely best characterizes the ethnic minority group, which is more predisposed to T2DM. The higher prevalence of T2DM in the SAS may be associated with the gut dysbiosis observed.
Collapse
Affiliation(s)
- Eric I Nayman
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.
- Bioinformatics Research Group, Knight Foundation School of Computing and Information Sciences, College of Engineering and Computing, Florida International University, Miami, FL, USA.
| | - Brooke A Schwartz
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Bioinformatics Research Group, Knight Foundation School of Computing and Information Sciences, College of Engineering and Computing, Florida International University, Miami, FL, USA
| | - Michaela Polmann
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Alayna C Gumabong
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Bioinformatics Research Group, Knight Foundation School of Computing and Information Sciences, College of Engineering and Computing, Florida International University, Miami, FL, USA
| | - Max Nieuwdorp
- Amsterdam Diabetes Center, Department of Internal Medicine, Academic Medical Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Trevor Cickovski
- Bioinformatics Research Group, Knight Foundation School of Computing and Information Sciences, College of Engineering and Computing, Florida International University, Miami, FL, USA.
| | - Kalai Mathee
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.
- Biomolecular Sciences Institute, Florida International University, Miami, FL, USA.
| |
Collapse
|
26
|
Zheng G, Wang D, Mao K, Wang M, Wang J, Xun W, Huang S. Exploring the Rumen Microbiota and Serum Metabolite Profile of Hainan Black Goats with Different Body Weights before Weaning. Animals (Basel) 2024; 14:425. [PMID: 38338068 PMCID: PMC10854652 DOI: 10.3390/ani14030425] [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: 12/28/2023] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
The critical role of the rumen microbiota in the growth performance of livestock is recognized, yet its significance in determining the body weight of goat kids before weaning remains less understood. To bridge this gap, our study delved into the rumen microbiota, serum metabolome, rumen fermentation, and rumen development in goat kids with contrasting body weights before weaning. We selected 10 goat kids from a cohort of 100, categorized into low body weight (LBW, 5.56 ± 0.98 kg) and high body weight (HBW, 9.51 ± 1.01 kg) groups. The study involved sampling rumen contents, tissues, and serum from these animals. Our findings showed that the HBW goat kids showed significant enrichment of VFA-producing bacteria, particularly microbiota taxa within the Prevotellaceae genera (UCG-001, UCG-003, and UCG-004) and the Prevotella genus. This enrichment correlated with elevated acetate and butyrate levels, positively influencing rumen papillae development. Additionally, it was associated with elevated serum levels of glucose, total cholesterol, and triglycerides. The serum metabonomic analysis revealed marked differences in fatty acid metabolism between the LBW and HBW groups, particularly in encompassing oleic acid and both long-chain saturated and polyunsaturated fatty acids. Further correlational analysis underscored a significant positive association between Prevotellaceae_UCG-001 and specific lipids, such as phosphatidylcholine (PC) (22:5/18:3) and PC (20:3/20:1) (r > 0.60, p < 0.05). In summary, this study underscores the pivotal role of the rumen microbiota in goat kids' weight and its correlation with specific serum metabolites. These insights could pave the way for innovative strategies aimed at improving animal body weight through targeted modulation of the rumen microbiota.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shuai Huang
- Forage Processing and Ruminant Nutrition Laboratory, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China; (G.Z.)
| |
Collapse
|
27
|
Tao Z, Wang Y. The health benefits of dietary short-chain fatty acids in metabolic diseases. Crit Rev Food Sci Nutr 2024; 65:1579-1592. [PMID: 38189336 DOI: 10.1080/10408398.2023.2297811] [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] [Indexed: 01/09/2024]
Abstract
Short-chain fatty acids (SCFAs) are a subset of fatty acids that play crucial roles in maintaining normal physiology and developing metabolic diseases, such as obesity, diabetes, cardiovascular disease, and liver disease. Even though dairy products and vegetable oils are the direct dietary sources of SCFAs, their quantities are highly restricted. SCFAs are produced indirectly through microbial fermentation of fibers. The biological roles of SCFAs in human health and metabolic diseases are mainly due to their receptors, GPR41 and GPR43, FFAR2 and FFAR3. Additionally, it has been demonstrated that SCFAs modulate DNMTs and HDAC activities, inhibit NF-κB-STAT signaling, and regulate G(i/o)βγ-PLC-PKC-PTEN signaling and PPARγ-UCP2-AMPK autophagic signaling, thus mitigating metabolic diseases. Recent studies have uncovered that SCFAs play crucial roles in epigenetic modifications of DNAs, RNAs, and post-translational modifications of proteins, which are critical regulators of metabolic health and diseases. At the same time, dietary recommendations for the purpose of SCFAs have been proposed. The objective of the review is to summarize the most recent research on the role of dietary SCFAs in metabolic diseases, especially the signal transduction of SCFAs in metabolic diseases and their functional efficacy in different backgrounds and models of metabolic diseases, at the same time, to provide dietary and nutritional recommendations for using SCFAs as food ingredients to prevent metabolic diseases.
Collapse
Affiliation(s)
- Zhipeng Tao
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
- Department of Nutrition Sciences, Texas Woman's University, Denton, Texas, USA
| | - Yao Wang
- Diabetes Center, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
28
|
Tuohy K, Vaughan EE, Harthoorn LF, Blaak EE, Burnet PWJ, Busetti A, Chakrabarti A, Delzenne N, de Vos P, Dye L, Guillemet D, Houghton LA, Kardinaal AFM, Mersh C, Musa-Veloso K, Nielsen A, Palasinska J, Salminen S, Walton G, Venlet N, Hubermont C, Calder PC. Prebiotics in food and dietary supplements: a roadmap to EU health claims. Gut Microbes 2024; 16:2428848. [PMID: 39544074 PMCID: PMC11572068 DOI: 10.1080/19490976.2024.2428848] [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: 06/14/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 11/17/2024] Open
Abstract
Numerous studies have established that prebiotic ingredients in foods and dietary supplements may play a role in supporting human health. Over the three decades that have passed since prebiotics were first defined as a concept, research has revealed a complex universe of prebiotic-induced changes to the human microbiota. There are strong indications of a direct link between these prebiotic-induced changes and specific health benefits. However, at the present time, the EU has not permitted use of the term 'prebiotic' in connection with an approved health claim. This paper is the outcome of a workshop organized on the 25th October 2023 by the European branch of the International Life Science Institute (ILSI). It provides an overview of the regulatory requirements for authorized health claims in the EU, key areas of prebiotic research, and findings to date in relation to prebiotics and digestive, immune, metabolic and cognitive health. Research gaps and documentation challenges are then explored and a roadmap proposed for achieving authorization of 'prebiotic' in the wording of future EU health claims.
Collapse
Affiliation(s)
- Kieran Tuohy
- School of Food Science & Nutrition, University of Leeds, Leeds, UK
| | | | | | - Ellen E. Blaak
- Department of Human Biology, NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | | | | | | | - Nathalie Delzenne
- Woluwe Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
| | - Paul de Vos
- Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Louise Dye
- Institute for Sustainable Food, University of Sheffield, Sheffield, UK
| | | | | | | | - Cath Mersh
- Cath Mersh Communications, Aarhus, Denmark
| | | | | | | | - Seppo Salminen
- Center for Food and Nutrition Research, Faculty of Medicine, University of Turku, Turku, Finland
| | - Gemma Walton
- Food and Nutritional Sciences, University of Reading, Reading, UK
| | | | | | - Philip C. Calder
- School of Human Development and Health, Faculty of Medicine, United Kingdom and NIHR Southampton Biomedical, Research Centre, University Hospital NHS Foundation Trust and University of Southampton, Southampton, UK
| |
Collapse
|
29
|
de Wit DF, Hanssen NMJ, Wortelboer K, Herrema H, Rampanelli E, Nieuwdorp M. Evidence for the contribution of the gut microbiome to obesity and its reversal. Sci Transl Med 2023; 15:eadg2773. [PMID: 37992156 DOI: 10.1126/scitranslmed.adg2773] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 09/27/2023] [Indexed: 11/24/2023]
Abstract
Obesity has become a worldwide pandemic affecting more than 650 million people and is associated with a high burden of morbidity. Alongside traditional risk factors for obesity, the gut microbiome has been identified as a potential factor in weight regulation. Although rodent studies suggest a link between the gut microbiome and body weight, human evidence for causality remains scarce. In this Review, we postulate that existing evidence remains to establish a contribution of the gut microbiome to the development of obesity in humans but that modified probiotic strains and supraphysiological dosages of microbial metabolites may be beneficial in combatting obesity.
Collapse
Affiliation(s)
- Douwe F de Wit
- Amsterdam UMC location University of Amsterdam, Experimental Vascular Medicine, 1105AZ Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, 1105AZ Amsterdam, Netherlands
| | - Nordin M J Hanssen
- Amsterdam UMC location University of Amsterdam, Experimental Vascular Medicine, 1105AZ Amsterdam, Netherlands
| | - Koen Wortelboer
- Amsterdam UMC location University of Amsterdam, Experimental Vascular Medicine, 1105AZ Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, 1105AZ Amsterdam, Netherlands
| | - Hilde Herrema
- Amsterdam UMC location University of Amsterdam, Experimental Vascular Medicine, 1105AZ Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, 1105AZ Amsterdam, Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105AZ Amsterdam, Netherlands
| | - Elena Rampanelli
- Amsterdam UMC location University of Amsterdam, Experimental Vascular Medicine, 1105AZ Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, 1105AZ Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, 1105AZ Amsterdam, Netherlands
| | - Max Nieuwdorp
- Amsterdam UMC location University of Amsterdam, Experimental Vascular Medicine, 1105AZ Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, 1105AZ Amsterdam, Netherlands
- Amsterdam UMC location Vrije Universiteit Medical Center, Department of Internal Medicine, Diabetes Center, 1105AZ Amsterdam, Netherlands
| |
Collapse
|
30
|
Freitas PLD, Barros MVC, Fróes RBL, França LM, Paes AMDA. Prebiotic effects of plant-derived (poly)phenols on host metabolism: Is there a role for short-chain fatty acids? Crit Rev Food Sci Nutr 2023; 63:12285-12293. [PMID: 35833476 DOI: 10.1080/10408398.2022.2100315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The gut microbiota has been extensively investigated during the last decade because of its effects on host neuroendocrine pathways and other processes. The imbalance between beneficial and pathogenic bacteria, known as dysbiosis, may be a determining predisposing factor for many noncommunicable chronic diseases, such as obesity, type 2 diabetes mellitus, metabolic syndrome, and Alzheimer's disease. On the other hand, interventions aiming to reestablish the balance between microbiota components have been suggested as potential preventive therapeutic strategies against these disorders. Among these interventions, dietary supplementation with (poly)phenols has been highlighted due to the modulatory effects exerted by those compounds on the gut microbiota. In addition, (poly)phenol consumption is associated with increased production of short-chain fatty acids (SCFAs), a set of microbial metabolites whose actions are ascribed to improving the abovementioned metabolic disorders. Thus, this review discusses the modulation of the gut microbiota by prebiotic (poly)phenols based on in vivo studies performed with isolated (poly)phenolic compounds, their interaction with the gut microbiota and the production of SCFAs in pursuit of the molecular mechanisms underlying the health effects of (poly)phenols on host metabolism.
Collapse
Affiliation(s)
- Perla Lopes de Freitas
- Laboratory of Experimental Physiology (LeFisio), Department of Physiological Sciences, Biological and Health Sciences Center, Federal University of Maranhão, São Luís, Brazil
- Health Sciences Graduate Program, Biological and Health Sciences Center, Federal University of Maranhão, São Luís, Brazil
| | - Marcus Vinicius Câmara Barros
- Laboratory of Experimental Physiology (LeFisio), Department of Physiological Sciences, Biological and Health Sciences Center, Federal University of Maranhão, São Luís, Brazil
| | - Rômulo Brênno Lopes Fróes
- Laboratory of Experimental Physiology (LeFisio), Department of Physiological Sciences, Biological and Health Sciences Center, Federal University of Maranhão, São Luís, Brazil
| | - Lucas Martins França
- Laboratory of Experimental Physiology (LeFisio), Department of Physiological Sciences, Biological and Health Sciences Center, Federal University of Maranhão, São Luís, Brazil
- Health Sciences Graduate Program, Biological and Health Sciences Center, Federal University of Maranhão, São Luís, Brazil
| | - Antonio Marcus de Andrade Paes
- Laboratory of Experimental Physiology (LeFisio), Department of Physiological Sciences, Biological and Health Sciences Center, Federal University of Maranhão, São Luís, Brazil
- Health Sciences Graduate Program, Biological and Health Sciences Center, Federal University of Maranhão, São Luís, Brazil
| |
Collapse
|
31
|
Chu Z, Hu Z, Luo Y, Zhou Y, Yang F, Luo F. Targeting gut-liver axis by dietary lignans ameliorate obesity: evidences and mechanisms. Crit Rev Food Sci Nutr 2023; 65:243-264. [PMID: 37870876 DOI: 10.1080/10408398.2023.2272269] [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] [Indexed: 10/24/2023]
Abstract
An imbalance between energy consumption and energy expenditure causes obesity. It is characterized by increased adipose accumulation and accompanied by chronic low-grade inflammation. Many studies have suggested that the gut microbiota of the host mediates the relationship between high-fat diet consumption and the development of obesity. Diet and nutrition of the body are heavily influenced by gut microbiota. The alterations in the microbiota in the gut may have effects on the homeostasis of the host's energy levels, systemic inflammation, lipid metabolism, and insulin sensitivity. The liver is an important organ for fat metabolism and gut-liver axis play important role in the fat metabolism. Gut-liver axis is a bidirectional relationship between the gut and its microbiota and the liver. As essential plant components, lignans have been shown to have different biological functions. Accumulating evidences have suggested that lignans may have lipid-lowering properties. Lignans can regulate the level of the gut microbiota and their metabolites in the host, thereby affecting signaling pathways related to fat synthesis and metabolism. These signaling pathways can make a difference in inhibiting fat accumulation, accelerating energy metabolism, affecting appetite, and inhibiting chronic inflammation. It will provide the groundwork for future studies on the lipid-lowering impact of lignans and the creation of functional meals based on those findings.
Collapse
Affiliation(s)
- Zhongxing Chu
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P.R. China
| | - Zuomin Hu
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P.R. China
| | - Yi Luo
- Department of Clinic Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, P.R. China
| | - Yaping Zhou
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P.R. China
| | - Feiyan Yang
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P.R. China
| | - Feijun Luo
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P.R. China
| |
Collapse
|
32
|
Blaak EE, Goossens GH. Metabolic phenotyping in people living with obesity: Implications for dietary prevention. Rev Endocr Metab Disord 2023; 24:825-838. [PMID: 37581871 PMCID: PMC10492670 DOI: 10.1007/s11154-023-09830-4] [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] [Accepted: 08/01/2023] [Indexed: 08/16/2023]
Abstract
Given the increasing number of people living with obesity and related chronic metabolic disease, precision nutrition approaches are required to increase the effectiveness of prevention strategies. This review addresses these approaches in different metabolic phenotypes (metabotypes) in obesity. Although obesity is typically associated with an increased cardiometabolic disease risk, some people with obesity are relatively protected against the detrimental effects of excess adiposity on cardiometabolic health, also referred to as 'metabolically healthy obesity' (MHO). Underlying mechanisms, the extent to which MHO is a transient state as well as lifestyle strategies to counteract the transition from MHO to metabolically unhealthy obesity (MUO) are discussed. Based on the limited resources that are available for dietary lifestyle interventions, it may be reasonable to prioritize interventions for people with MUO, since targeting high-risk patients for specific nutritional, lifestyle or weight-loss strategies may enhance the cost-effectiveness of these interventions. Additionally, the concept of tissue insulin resistant (IR) metabotypes is discussed, representing distinct etiologies towards type 2 diabetes (T2D) as well as cardiovascular disease (CVD). Recent evidence indicates that these tissue IR metabotypes, already present in individuals with obesity with a normal glucose homeostasis, respond differentially to diet. Modulation of dietary macronutrient composition according to these metabotypes may considerably improve cardiometabolic health benefits. Thus, nutritional or lifestyle intervention may improve cardiometabolic health, even with only minor or no weight loss, which stresses the importance of focusing on a healthy lifestyle and not on weight loss only. Targeting different metabotypes towards T2D and cardiometabolic diseases may lead to more effective lifestyle prevention and treatment strategies. Age and sex-related differences in tissue metabotypes and related microbial composition and functionality (fermentation), as important drivers and/or mediators of dietary intervention response, have to be taken into account. For the implementation of these approaches, more prospective trials are required to provide the knowledge base for precision nutrition in the prevention of chronic metabolic diseases.
Collapse
Affiliation(s)
- Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| |
Collapse
|
33
|
Ou H, Chen Q, Lin Z, Yang Y, Wang P, Sriboonvorakul N, Lin S. RNA-seq Analysis Reveals Potential Synergic Effects of Acetate and Cold Exposure on Interscapular Brown Adipose Tissue in Mice. BIOLOGY 2023; 12:1285. [PMID: 37886995 PMCID: PMC10603878 DOI: 10.3390/biology12101285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/22/2023] [Accepted: 09/24/2023] [Indexed: 10/28/2023]
Abstract
Brown adipose tissue (BAT) exhibits remarkable morphological and functional plasticity in response to environmental (e.g., cold exposure) and nutrient (e.g., high-fat diet) stimuli. Notably, a number of studies have showed that acetate, the main fermentation product of dietary fiber in gut, profoundly influences the differentiation and activity of BAT. However, the potential synergic or antagonistic effects of acetate and cold exposure on BAT have not been well examined. In the present study, the C57BL/6J mice were treated with acetate at the systemic level before a short period of cold exposure. Physiological parameters including body weight, blood glucose, and Respiratory Exchange Ratio (RER) were monitored, and thermal imaging of body surface temperature was captured. Moreover, the transcriptome profiles of interscapular BAT were also determined and analyzed afterwards. The obtained results showed that acetate treatment prior to cold exposure could alter the gene expression profile, as evidenced by significant differential clusters between the two groups. GO analysis and KEGG analysis further identified differentially expressed genes being mainly enriched for a number of biological terms and pathways related to lipid metabolism and brown adipose activity such as "G-protein-coupled receptor activity", "cAMP metabolic process", "PPAR signaling pathway", and "FoxO signaling pathway". GSEA analysis further suggested that activation status of key pathways including "PPAR signaling pathway" and "TCA cycle" were altered upon acetate treatment. Taken together, our study identified the potential synergistic effect of acetic acid with cold exposure on BAT, which highlighted the positive dietary and therapeutic aspects of acetate.
Collapse
Affiliation(s)
- Hongtao Ou
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (H.O.)
| | - Qingyan Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (H.O.)
| | - Zhongjing Lin
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (H.O.)
| | - Yang Yang
- Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen 518051, China
| | - Peixin Wang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (H.O.)
| | - Natthida Sriboonvorakul
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Shaoling Lin
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (H.O.)
| |
Collapse
|
34
|
Zhao G, Teng J, Dong R, Ban Q, Yang L, Du K, Wang Y, Pu H, Yang CS, Ren Z. Alleviating effects and mechanisms of action of large-leaf yellow tea drinking on diabetes and diabetic nephropathy in mice. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2023.02.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
35
|
Zhu M, Dagah OMA, Silaa BB, Lu J. Thioredoxin/Glutaredoxin Systems and Gut Microbiota in NAFLD: Interplay, Mechanism, and Therapeutical Potential. Antioxidants (Basel) 2023; 12:1680. [PMID: 37759983 PMCID: PMC10525532 DOI: 10.3390/antiox12091680] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/20/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common clinical disease, and its pathogenesis is closely linked to oxidative stress and gut microbiota dysbiosis. Recently accumulating evidence indicates that the thioredoxin and glutaredoxin systems, the two thiol-redox dependent antioxidant systems, are the key players in the NAFLD's development and progression. However, the effects of gut microbiota dysbiosis on the liver thiol-redox systems are not well clarified. This review explores the role and mechanisms of oxidative stress induced by bacteria in NAFLD while emphasizing the crucial interplay between gut microbiota dysbiosis and Trx mediated-redox regulation. The paper explores how dysbiosis affects the production of specific gut microbiota metabolites, such as trimethylamine N-oxide (TMAO), lipopolysaccharides (LPS), short-chain fatty acids (SCFAs), amino acids, bile acid, and alcohol. These metabolites, in turn, significantly impact liver inflammation, lipid metabolism, insulin resistance, and cellular damage through thiol-dependent redox signaling. It suggests that comprehensive approaches targeting both gut microbiota dysbiosis and the thiol-redox antioxidant system are essential for effectively preventing and treating NAFLD. Overall, comprehending the intricate relationship between gut microbiota dysbiosis and thiol-redox systems in NAFLD holds significant promise in enhancing patient outcomes and fostering the development of innovative therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | - Jun Lu
- Engineering Research Center of Coptis Development and Utilization/Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education (Southwest University), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China; (M.Z.); (O.M.A.D.); (B.B.S.)
| |
Collapse
|
36
|
Masse KE, Lu VB. Short-chain fatty acids, secondary bile acids and indoles: gut microbial metabolites with effects on enteroendocrine cell function and their potential as therapies for metabolic disease. Front Endocrinol (Lausanne) 2023; 14:1169624. [PMID: 37560311 PMCID: PMC10407565 DOI: 10.3389/fendo.2023.1169624] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/05/2023] [Indexed: 08/11/2023] Open
Abstract
The gastrointestinal tract hosts the largest ecosystem of microorganisms in the body. The metabolism of ingested nutrients by gut bacteria produces novel chemical mediators that can influence chemosensory cells lining the gastrointestinal tract. Specifically, hormone-releasing enteroendocrine cells which express a host of receptors activated by these bacterial metabolites. This review will focus on the activation mechanisms of glucagon-like peptide-1 releasing enteroendocrine cells by the three main bacterial metabolites produced in the gut: short-chain fatty acids, secondary bile acids and indoles. Given the importance of enteroendocrine cells in regulating glucose homeostasis and food intake, we will also discuss therapies based on these bacterial metabolites used in the treatment of metabolic diseases such as diabetes and obesity. Elucidating the mechanisms gut bacteria can influence cellular function in the host will advance our understanding of this fundamental symbiotic relationship and unlock the potential of harnessing these pathways to improve human health.
Collapse
Affiliation(s)
| | - Van B. Lu
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| |
Collapse
|
37
|
Canfora EE, Vliex LMM, Wang T, Nauta A, Bouwman FG, Holst JJ, Venema K, Zoetendal EG, Blaak EE. 2'-fucosyllactose alone or combined with resistant starch increases circulating short-chain fatty acids in lean men and men with prediabetes and obesity. Front Nutr 2023; 10:1200645. [PMID: 37529001 PMCID: PMC10388544 DOI: 10.3389/fnut.2023.1200645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/09/2023] [Indexed: 08/03/2023] Open
Abstract
Background Infusion of short-chain fatty acids (SCFA) to the distal colon beneficially affects human substrate and energy metabolism. Here, we hypothesized that the combination of 2'-fucosyllactose (2'-FL) with resistant starch (RS) increases distal colonic SCFA production and improves metabolic parameters. Methods In this randomized, crossover study, 10 lean (BMI 20-24.9 kg/m2) and nine men with prediabetes and overweight/obesity (BMI 25-35 kg/m2) were supplemented with either 2'-FL, 2'-FL+RS, or placebo one day before a clinical investigation day (CID). During the CID, blood samples were collected after a overnight fast and after intake of a liquid high-fat mixed meal to determine plasma SCFA (primary outcomes). Secondary outcomes were fasting and postprandial plasma insulin, glucose, free fatty acid (FFA), glucagon-like peptide-1, and peptide YY concentrations. In addition, fecal SCFA and microbiota composition, energy expenditure and substrate oxidation (indirect calorimetry), and breath hydrogen excretion were determined. Results In lean men, supplementation with 2'-FL increased postprandial plasma acetate (P = 0.017) and fasting H2 excretion (P = 0.041) compared to placebo. Postprandial plasma butyrate concentration increased after 2'-FL and 2'-FL+RS as compared to placebo (P < 0.05) in lean men and men with prediabetes and overweight/obesity. Additionally, 2'-FL+RS decreased fasting and postprandial plasma FFA concentrations compared to placebo (P < 0.05) in lean men. Conclusion Supplementation of 2'-FL with/without RS the day before investigation increased systemic butyrate concentrations in lean men as well as in men with prediabetes and obesity, while acetate only increased in lean men. The combination of 2'-FL with RS showed a putatively beneficial metabolic effect by lowering plasma FFA in lean men, indicating a phenotype-specific effect. Clinical trial registration nr. NCT04795804.
Collapse
Affiliation(s)
- Emanuel E. Canfora
- Human Biology, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, Netherlands
| | - Lars M. M. Vliex
- Human Biology, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, Netherlands
| | - Taojun Wang
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, Netherlands
| | | | - Freek G. Bouwman
- Human Biology, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, Netherlands
| | - Jens J. Holst
- NovoNordisk Center for Basic Metabolic Research and Department of Biomedical Sciences, Faculty of Health and Medical Sciences University of Copenhagen, Copenhagen, Denmark
| | - Koen Venema
- Maastricht University—Campus Venlo, Centre for Healthy Eating and Food Innovation, Venlo, Netherlands
| | - Erwin G. Zoetendal
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, Netherlands
| | - Ellen E. Blaak
- Human Biology, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
38
|
Wei B, Wang L, Su L, Tao X, Chen S, Wu J, Xia W. Structural characterization of slow digestion dextrin synthesized by a combination of α-glucosidase and cyclodextrin glucosyltransferase and its prebiotic potential on the gut microbiota in vitro. Food Chem 2023; 426:136554. [PMID: 37321121 DOI: 10.1016/j.foodchem.2023.136554] [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: 09/12/2022] [Revised: 05/16/2023] [Accepted: 06/04/2023] [Indexed: 06/17/2023]
Abstract
Starch-based dietary fibers are at the forefront of functional ingredient research. In this study, a novel water-soluble slow digestion dextrin (SDD) was synthesized by synergy of α-glucosidase and cyclodextrin glucosyltransferase and characterized. Results showed that SDD exhibited high solubility, low viscosity, and resistance to digestive enzymes, and also showed an increased dietary fiber content of 45.7% compared with that of α-glucosidase catalysis alone. Furthermore, SDD was used as the sole carbon source to ferment selected intestinal strains and human fecal microflora in vitro to investigate its prebiotic effects. It was found that SDD could markedly enriched the abundance of Bifidobacterium, Veillonella, Dialister, and Blautia in human gut microflora and yielded higher total organic acid. The combination of α-glucosidase and cyclodextrin glucosyltransferase in this study showed valuable potential for the preparation of a novel slow digestion dextrin with good physicochemical properties and improved prebiotic effects.
Collapse
Affiliation(s)
- Beibei Wei
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China; School of Biotechnology and Key Laboratory of Industrial Biotechnology Ministry of Education, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Lei Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China; School of Biotechnology and Key Laboratory of Industrial Biotechnology Ministry of Education, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Lingqia Su
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China; School of Biotechnology and Key Laboratory of Industrial Biotechnology Ministry of Education, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Xiumei Tao
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China; School of Biotechnology and Key Laboratory of Industrial Biotechnology Ministry of Education, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Sheng Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China; School of Biotechnology and Key Laboratory of Industrial Biotechnology Ministry of Education, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Jing Wu
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China; School of Biotechnology and Key Laboratory of Industrial Biotechnology Ministry of Education, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Wei Xia
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China; School of Biotechnology and Key Laboratory of Industrial Biotechnology Ministry of Education, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China.
| |
Collapse
|
39
|
Lange O, Proczko-Stepaniak M, Mika A. Short-Chain Fatty Acids-A Product of the Microbiome and Its Participation in Two-Way Communication on the Microbiome-Host Mammal Line. Curr Obes Rep 2023:10.1007/s13679-023-00503-6. [PMID: 37208544 DOI: 10.1007/s13679-023-00503-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/29/2023] [Indexed: 05/21/2023]
Abstract
PURPOSE OF REVIEW The review aims to describe short-chain fatty acids (SCFAs) as metabolites of bacteria, their complex influence on whole-body metabolism, and alterations in the SCFA profile in obesity and after bariatric surgery (BS). RECENT FINDINGS The fecal profile of SCFAs in obese patients differs from that of lean patients, as well as their gut microbiota composition. In obese patients, a lower diversity of bacteria is observed, as well as higher concentrations of SCFAs in stool samples. Obesity is now considered a global epidemic and bariatric surgery (BS) is an effective treatment for severe obesity. BS affects the structure and functioning of the digestive system, and also alters gut microbiota and the concentration of fecal SCFAs. Generally, after BS, SCFA levels are lower but levels of branched short-chain fatty acids (BSCFAs) are elevated, the effect of which is not fully understood. Moreover, changes in the profile of circulating SCFAs are little known and this is an area for further research. Obesity seems to be inherently associated with changes in the SCFA profile. It is necessary to better understand the impact of BS on microbiota and the metabolome in both feces and blood as only a small percentage of SCFAs are excreted. Further research may allow the development of a personalized therapeutic approach to the BS patient in terms of diet and prebiotic intervention.
Collapse
Affiliation(s)
- Oliwia Lange
- Department of Environmental Analysis, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland
| | - Monika Proczko-Stepaniak
- Department of General, Endocrine, and Transplant Surgery, Faculty of Medicine, Medical University of Gdansk, Smoluchowskiego 17, 80-214, Gdansk, Poland
| | - Adriana Mika
- Department of Environmental Analysis, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland.
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland.
| |
Collapse
|
40
|
Park SJ, Sharma A, Lee HJ. Postbiotics against Obesity: Perception and Overview Based on Pre-Clinical and Clinical Studies. Int J Mol Sci 2023; 24:6414. [PMID: 37047387 PMCID: PMC10095054 DOI: 10.3390/ijms24076414] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/13/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Overweight and obesity are significant global public health concerns that are increasing in prevalence at an alarming rate. Numerous studies have demonstrated the benefits of probiotics against obesity. Postbiotics are the next generation of probiotics that include bacteria-free extracts and nonviable microorganisms that may be advantageous to the host and are being increasingly preferred over regular probiotics. However, the impact of postbiotics on obesity has not been thoroughly investigated. Therefore, the goal of this review is to gather in-depth data on the ability of postbiotics to combat obesity. Postbiotics have been reported to have significant potential in alleviating obesity. This review comprehensively discusses the anti-obesity effects of postbiotics in cellular, animal, and clinical studies. Postbiotics exert anti-obesity effects via multiple mechanisms, with the major mechanisms including increased energy expenditure, reduced adipogenesis and adipocyte differentiation, suppression of food intake, inhibition of lipid absorption, regulation of lipid metabolism, and regulation of gut dysbiosis. Future research should include further in-depth studies on strain identification, scale-up of postbiotics, identification of underlying mechanisms, and well-defined clinical studies. Postbiotics could be a promising dietary intervention for the prevention and management of obesity.
Collapse
Affiliation(s)
- Seon-Joo Park
- Department of Food and Nutrition, College of Bionanotechnology, Gachon University, Seongnam-si 13120, Republic of Korea;
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam-si 13120, Republic of Korea
| | - Anshul Sharma
- Department of Food and Nutrition, College of Bionanotechnology, Gachon University, Seongnam-si 13120, Republic of Korea;
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of Bionanotechnology, Gachon University, Seongnam-si 13120, Republic of Korea;
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam-si 13120, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
41
|
Birkeland E, Gharagozlian S, Valeur J, Aas AM. Short-chain fatty acids as a link between diet and cardiometabolic risk: a narrative review. Lipids Health Dis 2023; 22:40. [PMID: 36915164 PMCID: PMC10012717 DOI: 10.1186/s12944-023-01803-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
AIM Diet has a profound impact on cardiometabolic health outcomes such as obesity, blood glucose, blood lipids and blood pressure. In recent years, the gut microbiota has emerged as one of several potential key players explaining dietary effects on these outcomes. In this review we aim to summarise current knowledge of interaction between diet and gut microbiota focusing on the gut-derived microbial metabolites short-chain fatty acids and their role in modulating cardiometabolic risk. FINDINGS Many observational and interventional studies in humans have found that diets rich in fibre or supplemented with prebiotic fibres have a favourable effect on the gut microbiota composition, with increased diversity accompanied by enhancement in short-chain fatty acids and bacteria producing them. High-fat diets, particularly diets high in saturated fatty acids, have shown the opposite effect. Several recent studies indicate that the gut microbiota modulates metabolic responses to diet in, e.g., postprandial blood glucose and blood lipid levels. However, the metabolic responses to dietary interventions, seem to vary depending on individual traits such as age, sex, ethnicity, and existing gut microbiota, as well as genetics. Studies mainly in animal models and cell lines have shown possible pathways through which short-chain fatty acids may mediate these dietary effects on metabolic regulation. Human intervention studies appear to support the favourable effect of short-chain fatty acid in animal studies, but the effects may be modest and vary depending on which cofactors were taken into consideration. CONCLUSION This is an expanding and active field of research that in the near future is likely to broaden our understanding of the role of the gut microbiota and short-chain fatty acids in modulating metabolic responses to diet. Nevertheless, the findings so far seem to support current dietary guidelines encouraging the intake of fibre rich plant-based foods and discouraging the intake of animal foods rich in saturated fatty acids.
Collapse
Affiliation(s)
- Eline Birkeland
- Section of Nutrition and Dietetics, Department of Clinical Service, Division of Medicine, Oslo University Hospital, Oslo, Norway
| | - Sedegheh Gharagozlian
- Section of Nutrition and Dietetics, Department of Clinical Service, Division of Medicine, Oslo University Hospital, Oslo, Norway
| | - Jørgen Valeur
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital, Oslo, Norway
| | - Anne-Marie Aas
- Section of Nutrition and Dietetics, Department of Clinical Service, Division of Medicine, Oslo University Hospital, Oslo, Norway. .,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
42
|
Bastings JJAJ, Venema K, Blaak EE, Adam TC. Influence of the gut microbiota on satiety signaling. Trends Endocrinol Metab 2023; 34:243-255. [PMID: 36870872 DOI: 10.1016/j.tem.2023.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/09/2023] [Accepted: 02/09/2023] [Indexed: 03/06/2023]
Abstract
Recent studies show a link between the gut microbiota and the regulation of satiety and energy intake, processes that contribute to the development and pathophysiology of metabolic diseases. However, this link is predominantly established in animal and in vitro studies, whereas human intervention studies are scarce. In this review we focus on recent evidence linking satiety and the gut microbiome, with specific emphasis on gut microbial short-chain fatty acids (SCFAs). Based on a systematic search we provide an overview of human studies linking the intake of prebiotics with gut microbial alterations and satiety signaling. Our outcomes highlight the importance of in-depth examination of the gut microbiota in relation to satiety and provide insights into recent and future studies in this field.
Collapse
Affiliation(s)
- Jacco J A J Bastings
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Koen Venema
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands; Centre for Healthy Eating and Food Innovation, Maastricht University, Campus Venlo, Venlo, The Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands.
| | - Tanja C Adam
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
43
|
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
|
44
|
van Deuren T, Smolders L, Hartog A, Bouwman FG, Holst JJ, Venema K, Blaak EE, Canfora EE. Butyrate and hexanoate-enriched triglycerides increase postprandrial systemic butyrate and hexanoate in men with overweight/obesity: A double-blind placebo-controlled randomized crossover trial. Front Nutr 2023; 9:1066950. [PMID: 36687671 PMCID: PMC9846253 DOI: 10.3389/fnut.2022.1066950] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/05/2022] [Indexed: 01/05/2023] Open
Abstract
Background Short chain fatty acids (SCFA) are increasingly recognized for their potential ability to alleviate obesity-associated chronic low-grade inflammation and disturbed energy homeostasis. Evidence suggests that an increase in circulating SCFA might be necessary to induce beneficial alterations in energy metabolism. Objective To compare the bioaccessibility of two different SCFA-enriched triglycerides: Akovita SCT (butyrate and hexanoate esterified with long chain fatty acids) and tributyrin/caproin (solely butyrate and hexanoate) and investigate whether the SCFA from orally administrated Akovita SCT reach the circulation and affect postprandial metabolism in men with overweight/obesity. Methods The site, speed, and amount of SCFA release from Akovita SCT and tributyrin/caproin were assessed in a validated In vitro Model of the stomach and small intestine (TIM-1). Subsequently, a double-blind placebo-controlled randomized crossover study was conducted at Maastricht University with fourteen men with overweight/obesity (BMI 25-35 kg/m2) of which twelve men finished all testdays and were included for analysis. The participants received a liquid high fat mixed meal test containing either a low (650 mg), medium (1,325 mg), or high dose (2,000 mg) of Akovita SCT or a placebo (sunflower oil) in randomized order. Blood was sampled at baseline and after ingestion for 6 h for the primary outcome plasma butyrate and hexanoate concentration. Secondary outcomes included hydrogen breath, appetite, gastrointestinal complaints, circulating glucagon-like peptide 1, free fatty acids, glucose, triglycerides, insulin, and cytokines concentrations. Results In TIM-1, tributyrin/caproin was rapidly cleaved in the gastric compartment whereas the release of SCFA from Akovita SCT occurred predominantly in the small intestine. In vivo, all doses were well-tolerated. The medium dose increased (P < 0.05) and the high dose tended to increase (P < 0.10) postprandial circulating butyrate and both doses increased circulating hexanoate (P < 0.05) compared to placebo. Nevertheless, Akovita SCT supplementation did not affect any secondary outcomes compared to placebo. Conclusion Esterifying SCFA-enriched triglycerides with long chain fatty acids delayed SCFA release from the glycerol backbone. Akovita SCT increased postprandial circulating butyrate and hexanoate without changing metabolic parameters in men with overweight/obesity. Future randomized clinical trials should investigate whether long-term Akovita SCT supplementation can aid in the treatment or prevention of metabolic disorders. Clinical trial registration www.ClinicalTrials.gov, identifier: NCT04662411.
Collapse
Affiliation(s)
- Thirza van Deuren
- Department of Human Biology, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, Netherlands
| | - Lotte Smolders
- AAK, Department of Biotechnology and Nutrition, AAK Netherlands BV, Zaandijk, Netherlands
| | - Anita Hartog
- AAK, Department of Biotechnology and Nutrition, AAK Netherlands BV, Zaandijk, Netherlands
| | - Freek G. Bouwman
- Department of Human Biology, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, Netherlands
| | - Jens J. Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Koen Venema
- Centre for Healthy Eating and Food Innovation, Maastricht University, Venlo, Netherlands
| | - Ellen E. Blaak
- Department of Human Biology, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, Netherlands
| | - Emanuel E. Canfora
- Department of Human Biology, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, Netherlands,*Correspondence: Emanuel E. Canfora ✉
| |
Collapse
|
45
|
Trouwborst I, Gijbels A, Jardon KM, Siebelink E, Hul GB, Wanders L, Erdos B, Péter S, Singh-Povel CM, de Vogel-van den Bosch J, Adriaens ME, Arts ICW, Thijssen DHJ, Feskens EJM, Goossens GH, Afman LA, Blaak EE. Cardiometabolic health improvements upon dietary intervention are driven by tissue-specific insulin resistance phenotype: A precision nutrition trial. Cell Metab 2023; 35:71-83.e5. [PMID: 36599304 DOI: 10.1016/j.cmet.2022.12.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/12/2022] [Accepted: 11/13/2022] [Indexed: 01/05/2023]
Abstract
Precision nutrition based on metabolic phenotype may increase the effectiveness of interventions. In this proof-of-concept study, we investigated the effect of modulating dietary macronutrient composition according to muscle insulin-resistant (MIR) or liver insulin-resistant (LIR) phenotypes on cardiometabolic health. Women and men with MIR or LIR (n = 242, body mass index [BMI] 25-40 kg/m2, 40-75 years) were randomized to phenotype diet (PhenoDiet) group A or B and followed a 12-week high-monounsaturated fatty acid (HMUFA) diet or low-fat, high-protein, and high-fiber diet (LFHP) (PhenoDiet group A, MIR/HMUFA and LIR/LFHP; PhenoDiet group B, MIR/LFHP and LIR/HMUFA). PhenoDiet group B showed no significant improvements in the primary outcome disposition index, but greater improvements in insulin sensitivity, glucose homeostasis, serum triacylglycerol, and C-reactive protein compared with PhenoDiet group A were observed. We demonstrate that modulating macronutrient composition within the dietary guidelines based on tissue-specific insulin resistance (IR) phenotype enhances cardiometabolic health improvements. Clinicaltrials.gov registration: NCT03708419, CCMO registration NL63768.068.17.
Collapse
Affiliation(s)
- Inez Trouwborst
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands; TI Food and Nutrition (TIFN), Wageningen, the Netherlands
| | - Anouk Gijbels
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Kelly M Jardon
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands; TI Food and Nutrition (TIFN), Wageningen, the Netherlands
| | - Els Siebelink
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Gabby B Hul
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands; TI Food and Nutrition (TIFN), Wageningen, the Netherlands
| | - Lisa Wanders
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Radboud Institute for Health Sciences, Department of Physiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Balázs Erdos
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Maastricht Centre for Systems Biology, Maastricht University, Maastricht, the Netherlands
| | | | | | | | - Michiel E Adriaens
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Maastricht Centre for Systems Biology, Maastricht University, Maastricht, the Netherlands
| | - Ilja C W Arts
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Maastricht Centre for Systems Biology, Maastricht University, Maastricht, the Netherlands
| | - Dick H J Thijssen
- Radboud Institute for Health Sciences, Department of Physiology, Radboud University Medical Center, Nijmegen, the Netherlands; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Edith J M Feskens
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Lydia A Afman
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands; TI Food and Nutrition (TIFN), Wageningen, the Netherlands.
| |
Collapse
|
46
|
Manoharan N, Parasuraman R, Jayamurali D, Govindarajulu SN. The therapeutic role of microbial metabolites in human health and diseases. RECENT ADVANCES AND FUTURE PERSPECTIVES OF MICROBIAL METABOLITES 2023:1-38. [DOI: 10.1016/b978-0-323-90113-0.00002-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
47
|
Zhong Y, Wang T, Luo R, Liu J, Jin R, Peng X. Recent advances and potentiality of postbiotics in the food industry: Composition, inactivation methods, current applications in metabolic syndrome, and future trends. Crit Rev Food Sci Nutr 2022; 64:5768-5792. [PMID: 36537328 DOI: 10.1080/10408398.2022.2158174] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Postbiotics are defined as "preparation of inanimate microorganisms and/or their components that confers a health benefit on the host". Postbiotics have unique advantages over probiotics, such as stability, safety, and wide application. Although postbiotics are research hotspots, the research on them is still very limited. This review provides comprehensive information on the scope of postbiotics, the preparation methods of inanimate microorganisms, and the application and mechanisms of postbiotics in metabolic syndrome (MetS). Furthermore, the application trends of postbiotics in the food industry are reviewed. It was found that postbiotics mainly include inactivated microorganisms, microbial lysates, cell components, and metabolites. Thermal treatments are the main methods to prepare inanimate microorganisms as postbiotics, while non-thermal treatments, such as ionizing radiation, ultraviolet light, ultrasound, and supercritical CO2, show great potential in postbiotic preparation. Postbiotics could ameliorate MetS through multiple pathways including the modulation of gut microbiota, the enhancement of intestinal barrier, the regulation of inflammation and immunity, and the modulation of hormone homeostasis. Additionally, postbiotics have great potential in the food industry as functional food supplements, food quality improvers, and food preservatives. In addition, the SWOT analyses showed that the development of postbiotics in the food industry exists both opportunities and challenges.
Collapse
Affiliation(s)
- Yujie Zhong
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Tao Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan Province, China
| | - Ruilin Luo
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Jiayu Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Ruyi Jin
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaoli Peng
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
48
|
Zhang YM, Zhao YB, Li CQ, Wang L, Tian F, Erdene K, Ao CJ, Jin H. Relationships between rumen microbes, short-chain fatty acids, and markers of white adipose tissue browning during the cold season in grazing Mongolian sheep (Ovis aries). J Therm Biol 2022; 110:103386. [DOI: 10.1016/j.jtherbio.2022.103386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/20/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
|
49
|
Atanasova-Panchevska N, Stojchevski R, Hadzi-Petrushev N, Mitrokhin V, Avtanski D, Mladenov M. Antibacterial and Antiviral Properties of Tetrahydrocurcumin-Based Formulations: An Overview of Their Metabolism in Different Microbiotic Compartments. Life (Basel) 2022; 12:1708. [PMID: 36362863 PMCID: PMC9696410 DOI: 10.3390/life12111708] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/13/2022] [Accepted: 10/23/2022] [Indexed: 08/29/2023] Open
Abstract
In this review, the basic metabolic characteristics of the curcuminoid tetrahydrocurcumin (THC) at the level of the intestinal microbiota were addressed. Special attention was given to the bactericidal effects of one of the THC-phospholipid formulations, which has shown greater bioavailability and activity than pure THC. Similarly, quinoline derivatives and amino acid conjugates of THC have also shown antibacterial effects in the gut. The microbial effect of pure THC is particularly pronounced in pathophysiological conditions related to the function of the intestinal microbiota, such as type II diabetes. Furthermore, the antiviral characteristics of Cur compared to those of THC are more pronounced in preventing the influenza virus. In the case of HIV infections, the new microemulsion gel formulations of THC possess high retention during preventive application in the vagina and, at the same time, do not disturb the vaginal microbiota, which is critical in maintaining low vaginal pH. Based on the reviewed literature, finding new formulations of THC which can increase its bioavailability and activity and emphasize its antibacterial and antiviral characteristics could be very important. Applying such THC formulations in preventing and treating ailments related to the microbiotic compartments in the body would be beneficial from a medical point of view.
Collapse
Affiliation(s)
- Natalija Atanasova-Panchevska
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, P.O. Box 162, 1000 Skopje, North Macedonia
| | - Radoslav Stojchevski
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, P.O. Box 162, 1000 Skopje, North Macedonia
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, 110 E 59th Street, New York, NY 10022, USA
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, P.O. Box 162, 1000 Skopje, North Macedonia
| | - Vadim Mitrokhin
- Department of Physiology, Pirogov Russian National Research Medical University, Ostrovityanova Street, 1, 117997 Moscow, Russia
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, 110 E 59th Street, New York, NY 10022, USA
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, P.O. Box 162, 1000 Skopje, North Macedonia
- Department of Physiology, Pirogov Russian National Research Medical University, Ostrovityanova Street, 1, 117997 Moscow, Russia
| |
Collapse
|
50
|
van Deuren T, Blaak EE, Canfora EE. Butyrate to combat obesity and obesity-associated metabolic disorders: Current status and future implications for therapeutic use. Obes Rev 2022; 23:e13498. [PMID: 35856338 PMCID: PMC9541926 DOI: 10.1111/obr.13498] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/04/2022] [Accepted: 06/28/2022] [Indexed: 12/17/2022]
Abstract
Evidence is increasing that disturbances in the gut microbiome may play a significant role in the etiology of obesity and type 2 diabetes. The short chain fatty acid butyrate, a major end product of the bacterial fermentation of indigestible carbohydrates, is reputed to have anti-inflammatory properties and positive effects on body weight control and insulin sensitivity. However, whether butyrate has therapeutic potential for the treatment and prevention of obesity and obesity-related complications remains to be elucidated. Overall, animal studies strongly indicate that butyrate administered via various routes (e.g., orally) positively affects adipose tissue metabolism and functioning, energy and substrate metabolism, systemic and tissue-specific inflammation, and insulin sensitivity and body weight control. A limited number of human studies demonstrated interindividual differences in clinical effectiveness suggesting that outcomes may depend on the metabolic, microbial, and lifestyle-related characteristics of the target population. Hence, despite abundant evidence from animal data, support of human data is urgently required for the implementation of evidence-based oral and gut-derived butyrate interventions. To increase the efficacy of butyrate-focused interventions, future research should investigate which factors impact treatment outcomes including baseline gut microbial activity and functionality, thereby optimizing targeted-interventions and identifying individuals that merit most from such interventions.
Collapse
Affiliation(s)
- Thirza van Deuren
- Department of Human Biology, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Ellen E Blaak
- Department of Human Biology, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Emanuel E Canfora
- Department of Human Biology, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|