1
|
Delzenne NM, Bindels LB, Neyrinck AM, Walter J. The gut microbiome and dietary fibres: implications in obesity, cardiometabolic diseases and cancer. Nat Rev Microbiol 2025; 23:225-238. [PMID: 39390291 DOI: 10.1038/s41579-024-01108-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 10/12/2024]
Abstract
Dietary fibres constitute a heterogeneous class of nutrients that are key in the prevention of various chronic diseases. Most dietary fibres are fermented by the gut microbiome and may, thereby, modulate the gut microbial ecology and metabolism, impacting human health. Dietary fibres may influence the occurrence of specific bacterial taxa, with this effect varying between individuals. The effect of dietary fibres on microbial diversity is a matter of debate. Most intervention studies with dietary fibres in the context of obesity and related metabolic disorders reveal the need for an accurate assessment of the microbiome to better understand the variable response to dietary fibres. Epidemiological studies confirm that a high dietary fibre intake is strongly associated with a reduced occurrence of many types of cancer. However, there is a need to determine the impact of intervention with specific dietary fibres on cancer risk, therapy efficacy and toxicity, as well as in cancer cachexia. In this Review, we summarize the mechanisms by which the gut microbiome can mediate the physiological benefits of dietary fibres in the contexts of obesity, cardiometabolic diseases and cancer, their incidence being clearly linked to low dietary fibre intake.
Collapse
Affiliation(s)
- Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium.
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Jens Walter
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
2
|
Amadieu C, Ahmed H, Leclercq S, Koistinen V, Leyrolle Q, Stärkel P, Bindels LB, Layé S, Neyrinck AM, Kärkkäinen O, De Timary P, Hanhineva K, Delzenne NM. Effect of inulin supplementation on fecal and blood metabolome in alcohol use disorder patients: A randomised, controlled dietary intervention. Clin Nutr ESPEN 2025; 66:361-371. [PMID: 39864520 DOI: 10.1016/j.clnesp.2025.01.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND AND AIMS Alcohol Use Disorder (AUD) is a psychiatric disorder characterized notably by gut microbial dysbiosis and insufficient dietary fiber (DF) intake. This study aims to investigate the effect of DF placebo-controlled intervention in patients suffering from AUD during a three-week period of alcohol withdrawal, in order to discover microbial-derived metabolites that could be involved in metabolic and behavioral status. METHODS A randomized, double-blind, placebo-controlled study was performed with 50 AUD patients supplemented with inulin (prebiotic DF) or maltodextrin (placebo) during 17 days. Fecal microbiota composition, plasma and fecal metabolomics (liquid chromatography coupled to mass spectrometry), blood markers of inflammation and hepatic alterations, and psychological assessment (questionnaires) were analyzed before and after the intervention. RESULTS Fecal metabolomics revealed 14 metabolites significantly modified by inulin versus placebo treatment (increased N8-acetylspermidine and decreased indole-3-butyric acid, 5-amino valeric acid betaine (5-AVAB) and bile acids). Thirteen plasma metabolites differentiated both treatments (higher levels of long-chain fatty acids, medium-chain acylcarnitines and sphingomyelin species, and reduced 3-methylhistidine by inulin versus placebo). Fecal Lachnoclostridium correlated with 6 of the identified fecal metabolites, whereas plasma lipidic moieties positively correlated with fecal Ruminococcus torques group and Flavonifractor. Interestingly, parameters reflecting liver alterations inversely correlated with sphingomyelin (SM 36:2). CONCLUSIONS Three weeks of inulin supplementation during alcohol withdrawal leads to specific and different changes in the plasma and fecal metabolome of AUD patients, some of these gut microbiota-related metabolites being correlated with liver function. TRIAL REGISTRATION NCT03803709, https://clinicaltrials.gov/ct2/show/NCT03803709.
Collapse
Affiliation(s)
- Camille Amadieu
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium; Université de Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, F-33000 Bordeaux, France
| | - Hany Ahmed
- Food Sciences Unit, Department of Life Technologies, University of Turku, Turku, Finland
| | - Sophie Leclercq
- Laboratory of Nutritional Psychiatry, Institute of Neuroscience, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Ville Koistinen
- Food Sciences Unit, Department of Life Technologies, University of Turku, Turku, Finland; School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Quentin Leyrolle
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium; Université de Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, F-33000 Bordeaux, France
| | - Peter Stärkel
- Department of Gastro-enterology, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium; WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Sophie Layé
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, F-33000 Bordeaux, France
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Olli Kärkkäinen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Philippe De Timary
- Department of Adult Psychiatry, Cliniques Universitaires Saint-Luc and Institute of Neuroscience, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Kati Hanhineva
- Food Sciences Unit, Department of Life Technologies, University of Turku, Turku, Finland; School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
3
|
Saeed H, Díaz LA, Gil-Gómez A, Burton J, Bajaj JS, Romero-Gomez M, Arrese M, Arab JP, Khan MQ. Microbiome-centered therapies for the management of metabolic dysfunction-associated steatotic liver disease. Clin Mol Hepatol 2025; 31:S94-S111. [PMID: 39604327 PMCID: PMC11925441 DOI: 10.3350/cmh.2024.0811] [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: 09/13/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a significant global health issue, affecting over 30% of the population worldwide due to the rising prevalence of metabolic risk factors such as obesity and type 2 diabetes mellitus. This spectrum of liver disease ranges from isolated steatosis to more severe forms such as steatohepatitis, fibrosis, and cirrhosis. Recent studies highlight the role of gut microbiota in MASLD pathogenesis, showing that dysbiosis significantly impacts metabolic health and the progression of liver disease. This review critically evaluates current microbiome-centered therapies in MASLD management, including prebiotics, probiotics, synbiotics, fecal microbiota transplantation, and emerging therapies such as engineered bacteria and bacteriophage therapy. We explore the scientific rationale, clinical evidence, and potential mechanisms by which these interventions influence MASLD. The gut-liver axis is crucial in MASLD, with notable changes in microbiome composition linked to disease progression. For instance, specific microbial profiles and reduced alpha diversity are associated with MASLD severity. Therapeutic strategies targeting the microbiome could modulate disease progression by improving gut permeability, reducing endotoxin-producing bacteria, and altering bile acid metabolism. Although promising, these therapies require further research to fully understand their mechanisms and optimize their efficacy. This review integrates findings from clinical trials and experimental studies, providing a comprehensive overview of microbiome-centered therapies' potential in managing MASLD. Future research should focus on personalized strategies, utilizing microbiome features, blood metabolites, and customized dietary interventions to enhance the effectiveness of these therapies.
Collapse
Affiliation(s)
- Huma Saeed
- Division of Infectious Diseases, Department of Medicine, University of Western Ontario, London, ON, Canada
| | - Luis Antonio Díaz
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, CA, USA
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Antonio Gil-Gómez
- SeLiver Group, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Jeremy Burton
- Department of Microbiology & Immunology, Western University, London, ON, Canada
| | - Jasmohan S Bajaj
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Manuel Romero-Gomez
- SeLiver Group, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- UCM Digestive diseases, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Marco Arrese
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Pablo Arab
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Mohammad Qasim Khan
- Division of Gastroenterology, Department of Medicine, University of Western Ontario, London, ON, Canada
- Department of Epidemiology and Biostatistics, University of Western Ontario, London, ON, Canada
| |
Collapse
|
4
|
Liu YF, Liu YY, Xiao Y, Huang WJ, Sun RX, Hu J, Fu XZ, Tian CX, Fu Q, Zhao JX. Shenlian Decoction Ameliorates LPS-Related Inflammation in db/db Mice: Coupling Network Pharmacology With Experimental Verification. J Diabetes Res 2025; 2025:3823051. [PMID: 39810933 PMCID: PMC11729506 DOI: 10.1155/jdr/3823051] [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: 07/06/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025] Open
Abstract
Background: Shenlian (SL) decoction, a renowned traditional Chinese formula for diabetes mellitus, has also been employed to treat intestinal disorders. Previous studies have demonstrated the efficacy of SL decoction in regulating blood glucose and intestinal bacteria. Nevertheless, further analysis is required to elucidate the mechanistic link between SL decoction-mediated improvement of intestinal function and treatment of Type 2 diabetes mellitus (T2DM). Methods: Firstly, the active ingredients of SL decoction were sourced from the Traditional Chinese Medicine System Pharmacology (TCMSP) database, with putative targets of active ingredients being predicted using the same database. Secondly, the Online Mendelian Inheritance in Man (OMIM) and GeneCards databases were employed to screen the aforementioned targets that act on T2DM, and protein-protein interaction (PPI) networks were constructed in accordance with the results. Thirdly, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted using the Database for Annotation, Visualization, and Integrated Discovery (DAVID), which resulted in a comprehensive analysis of the association between SL decoction for the treatment of T2DM and the modulation of intestinal functions. Finally, the effect of the SL decoction on predicted lipopolysaccharide (LPS)-related targets, as well as intestinal function markers, was validated through in vivo experimentation. Results: A total of 36 active ingredients and 145 potential targets of SL decoction were predicted. GO enrichment analysis indicated that the principal biological processes by which the SL decoction acted against T2DM were responses to LPSs, while KEGG enrichment analysis identified the nuclear factor kappa B (NF-κB) signaling pathway and toll-like receptor signaling pathway as the key pathways involved. The in vivo experiments showed that SL decoction improved glycolipid metabolism indexes, inflammatory factor levels, and LPS levels in db/db mice. The immunohistochemical results demonstrated that the SL decoction restored the expression of Occludin, Claudin-1, and ZO-1 in the intestine and inhibited the expression of toll-like receptor 4 (TLR4), myeloid differentiation primary response gene 88 (MYD88), and NF-κB in both the intestine and pancreas. Furthermore, it may influence the levels of short-chain fatty acids (SCFAs) in feces. Conclusions: This research investigated the multigene pharmacological mechanism of SL decoction against T2DM using network pharmacology and in vivo experiments. SL decoction treatment of T2DM may reverse inflammation by inhibiting LPS-related pathway activation and improving intestinal function.
Collapse
Affiliation(s)
- Yi-fan Liu
- Section II of Endocrinology & Nephropathy Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuan-yuan Liu
- Institute of Chinese Medical Literature and Culture, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yao Xiao
- Nephropathy Department, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wei-jun Huang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Rui-xi Sun
- Institute of Basic Theory, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Hu
- Section II of Endocrinology & Nephropathy Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao-zhe Fu
- Section II of Endocrinology & Nephropathy Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Chu-xiao Tian
- Section II of Endocrinology & Nephropathy Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qiang Fu
- Section II of Endocrinology & Nephropathy Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jin-xi Zhao
- Section II of Endocrinology & Nephropathy Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
5
|
Xiao Y, Huang L, Zhao J, Chen W, Lu W. The gut core microbial species Bifidobacterium longum: Colonization, mechanisms, and health benefits. Microbiol Res 2025; 290:127966. [PMID: 39547052 DOI: 10.1016/j.micres.2024.127966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 10/27/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024]
Abstract
Bifidobacterium longum (B. longum) is a species of the core microbiome in the human gut, whose abundance is closely associated with host age and health status. B. longum has been shown to modulate host gut microecology and have the potential to alleviate various diseases. Comprehensive understanding on the colonization mechanism of B. longum and mechanism of the host-B. longum interactions, can provide us possibility to prevent and treat human diseases through B. longum-directed strategies. In this review, we summarized the gut colonization characteristics of B. longum, discussed the diet factors that have ability/potential to enrich indigenous and/or ingested B. longum strains, and reviewed the intervention mechanisms of B. longum in multiple diseases. The key findings are as follows: First, B. longum has specialized colonization mechanisms, like a wide carbohydrate utilization spectrum that allows it to adapt to the host's diet, species-level conserved genes encoding bile salt hydrolase (BSHs), and appropriate bacterial surface structures. Second, dietary intervention (e.g., anthocyanins) could effectively improve the gut colonization of B. longum, demonstrating the feasibility of diet-tuned strain colonization. Finally, we analyzed the skewed abundance of B. longum in different types of diseases and summarized the main mechanisms by which B. longum alleviates digestive (repairing the intestinal mucosal barrier by stimulating Paneth cell activity), immune (up-regulating the regulatory T cell (Treg) populations and maintaining the balance of Th1/Th2), and neurological diseases (regulating the kynurenine pathway and quinolinic acid levels in the brain through the gut-brain axis).
Collapse
Affiliation(s)
- Yue Xiao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China.
| | - Lijuan Huang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China
| |
Collapse
|
6
|
Feng Y, Jin Q, Liu X, Lin T, Johnson A, Huang H. Advances in understanding dietary fiber: Classification, structural characterization, modification, and gut microbiome interactions. Compr Rev Food Sci Food Saf 2025; 24:e70092. [PMID: 39840651 PMCID: PMC11752078 DOI: 10.1111/1541-4337.70092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/19/2024] [Accepted: 11/29/2024] [Indexed: 01/23/2025]
Abstract
Gut microbiota and their metabolites profoundly impact host physiology. Targeted modulation of gut microbiota has been a long-term interest in the scientific community. Numerous studies have investigated the feasibility of utilizing dietary fibers (DFs) to modulate gut microbiota and promote the production of health-beneficial bacterial metabolites. However, the complexity of fiber structures, microbiota composition, and their dynamic interactions have hindered the precise prediction of the impact of DF on the gut microbiome. We address this issue with a new perspective, focusing on the inherent chemical and structural complexity of DFs and their interaction with gut microbiota. The chemical and structural complexity of fibers was thoroughly elaborated, encompassing the fibers' molecular composition, polymorphism, mesoscopic structures, porosity, and particle size. Advanced characterization techniques to investigate fiber structural properties were discussed. Additionally, we examined the interactions between DFs and gut microbiota. Finally, we summarized processing techniques to modify fiber structures for improving the fermentability of DF by gut microbiota. The structure of fibers, such as their crystallinity, porosity, degree of branching, and pore wettability, significantly impacts their interactions with gut microbiota. These structural differences also substantially affect fiber's fermentability and capability to modulate the composition of gut microbiota. Conventional approaches are not capable of investigating complex fiber properties and their influences on the gut microbiome; therefore, it is of the essence to involve advanced material characterization techniques and artificial intelligence to unveil more comprehensive information on this topic.
Collapse
Affiliation(s)
- Yiming Feng
- Department of Food Science and TechnologyVirginia TechBlacksburgVirginiaUSA
- Department of Biological Systems EngineeringVirginia TechBlacksburgVirginiaUSA
| | - Qing Jin
- Department of Food Science and TechnologyVirginia TechBlacksburgVirginiaUSA
- School of Food and AgricultureUniversity of MaineOronoMaineUSA
| | - Xuanbo Liu
- Department of Food Science and TechnologyVirginia TechBlacksburgVirginiaUSA
| | - Tiantian Lin
- Department of Food Science and TechnologyVirginia TechBlacksburgVirginiaUSA
| | - Andrea Johnson
- Department of Food Science and TechnologyVirginia TechBlacksburgVirginiaUSA
| | - Haibo Huang
- Department of Food Science and TechnologyVirginia TechBlacksburgVirginiaUSA
| |
Collapse
|
7
|
An R, Zhou X, Zhang J, Lyu C, Wang D. Responses of intestinal microbiota to inulin was initial microbiota context dependent and affected by the supplementation dosage. Food Res Int 2025; 200:115498. [PMID: 39779139 DOI: 10.1016/j.foodres.2024.115498] [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: 06/23/2024] [Revised: 11/15/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025]
Abstract
Intestinal microbiota could respond to dietary fibres that are fermented by the gut microbiota, like prebiotics. Nevertheless, the dynamics of intestinal microbial community longitudinally after prebiotics intake, are still largely unknown. The current study unrevealed the successional process of intestinal microbial community after inulin supplementation, and the effect of supplementation dosage thereof, based on analysis of 16S rRNA gene sequences in C57BL/6 mice. We found that independent of supplementation dosage, intake of inulin could affect the intestinal microbial community within a day. Thereafter, the intestinal microbial community kept evolving until the last day of the supplementation (day 14) as a successional process, which was represented by the succession between intermediate and sluggish inulin responders. Remarkably, the successional process was initial microbial community context dependent and affected by the supplementation dosage. Specifically, the supplementation dosage affected the successional speed and the composition of the intermediate and sluggish inulin responders. Decreasing the relative abundance of previously identified intermediate responders, altered the successional process during inulin supplementation. Collectively, independent of supplementation dosage, the response of intestinal microbial community was rapid and the inulin induced temporal dynamics was represented by the succession between the intermediate and sluggish inulin responders. Nevertheless, the inulin induced successional process was initial microbial community context dependent and affected by the supplementation dosage. Findings of the current study would aid in the understanding of intestinal microbes' assembly during inulin supplementation and provide valuable support for dietary recommendations regarding to the use of prebiotics from the intestinal microbiota point of view.
Collapse
Affiliation(s)
- Ran An
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Xilong Zhou
- State Key Laboratory of Dairy Biotechnology, Dairy Research Institute, Bright Dairy and Food, Shanghai, China
| | - Jing Zhang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Chenang Lyu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Dapeng Wang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
8
|
Visuthranukul C, Sriswasdi S, Tepaamorndech S, Chamni S, Leelahavanichkul A, Joyjinda Y, Aksornkitti V, Chomtho S. Enhancing gut microbiota and microbial function with inulin supplementation in children with obesity. Int J Obes (Lond) 2024; 48:1696-1704. [PMID: 39033197 PMCID: PMC11584386 DOI: 10.1038/s41366-024-01590-8] [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: 03/29/2024] [Revised: 06/24/2024] [Accepted: 07/10/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND AND OBJECTIVES Gut dysbiosis that resulted from the alteration between host-microbe interaction might worsen obesity-induced systemic inflammation. Gut microbiota manipulation by supplementation of prebiotic inulin may reverse metabolic abnormalities and improve obesity. This study aimed to determine whether inulin supplementation improved intestinal microbiota and microbial functional pathways in children with obesity. METHODS Children with obesity whose BMI above median + 2SDs were recruited to a randomized, double-blinded placebo-controlled study. The participants aged 7-15 years were assigned to inulin supplement extracted from Thai Jerusalem artichoke (intervention), maltodextrin (placebo), and dietary fiber advice groups. All participants received similar monthly conventional advice and follow-up for 6 months. Fecal samples were collected for gut microbiome analysis using 16S rRNA sequencing. Phylogenetic Investigation of Communities by Reconstruction of Unobserved States was performed to infer microbial functional pathways. RESULTS One hundred and forty-three children with available taxonomic and functional pathway abundance profiles were evaluated. A significant increase in alpha-diversity was observed in the inulin group. Inulin supplementation substantially enhanced Bifidobacterium, Blautia, Megasphaera, and several butyrate-producing bacteria, including Agathobacter, Eubacterium coprostanoligenes, and Subdoligranulum, compared to the other groups. The inulin group showed a significant difference in functional pathways of proteasome and riboflavin metabolism. These changes correlated with clinical and metabolic outcomes exclusively in the inulin group. CONCLUSIONS Inulin supplementation significantly promoted gut bacterial diversity and improved gut microbiota dysbiosis in children with obesity. The modulation of functional pathways by inulin suggests its potential to establish beneficial interactions between the gut microbiota and host physiology. Inulin supplementation could be a strategic treatment to restore the balance of intestinal microbiota and regulate their functions in childhood obesity.
Collapse
Affiliation(s)
- Chonnikant Visuthranukul
- Center of Excellence in Pediatric Nutrition, Division of Nutrition, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Sira Sriswasdi
- Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Computational Molecular Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Surapun Tepaamorndech
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Supakarn Chamni
- Center of Excellent in Natural Products and Nanoparticles (NP2), Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Inflammation and Immunology Research Unit (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Yutthana Joyjinda
- WHO-CC for Research and Training on Viral Zoonoses, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Thai Red Cross Emerging Infection Diseases-Health Science Center, Bangkok, 10330, Thailand
| | - Vitavat Aksornkitti
- Center of Excellence in Computational Molecular Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sirinuch Chomtho
- Center of Excellence in Pediatric Nutrition, Division of Nutrition, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| |
Collapse
|
9
|
Schwenger KJP, Ghorbani Y, Bharatselvam S, Chen L, Chomiak KM, Tyler AC, Eddingsaas NC, Fischer SE, Jackson TD, Okrainec A, Allard JP. Links between fecal microplastics and parameters related to metabolic dysfunction-associated steatotic liver disease (MASLD) in humans: An exploratory study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 953:176153. [PMID: 39260480 DOI: 10.1016/j.scitotenv.2024.176153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/06/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
Microplastics (MPs) can persist in the environment and human body. Murine studies showed that exposure to MPs could cause metabolic dysregulation, contributing metabolic dysfunction-associated steatotic liver disease (MASLD) or steatohepatitis (MASH). However, research on the role of MPs in humans is limited. Thus, we aimed to assess links between human fecal MPs and liver histology, gene expression, immune cells and intestinal microbiota (IM). We included 6 lean healthy liver donors and 6 normal liver (obese) and 11 MASH patients. Overall, pre-BSx, we observed no significant differences in fecal MPs between groups. However, fecal MP fibers and total MPs positively correlated with portal and total macrophages and total killer T cells while total fecal MPs were positively correlated with natural killer cells. Additionally, 19 genes related to immune system and apoptosis correlated with fecal MPs at baseline. Fecal MP fibers correlated positively with fecal Bifidobacterium and negatively with Lachnospiraceae. Patients with MASH (n = 11) were re-assessed 12-months post-bariatric surgery (BSx) and we found that those with persistent disease (n = 4) had higher fecal MP fragments than those with normalized liver histology (n = 7). At 12-month post-BSx, MP fragments positively correlated with helper T cells and total MPs positively correlated with natural killer T cells and B cells. Our study is the first to look at 1) the role of MPs in MASH and its association with IM, immune cells and hepatic gene expression and 2) look at the role of MPs longitudinally in MASH persistence following BSx. Future research should further explore this relationship.
Collapse
Affiliation(s)
| | - Yasaman Ghorbani
- Toronto General Hospital, University Health Network, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada
| | | | - Lina Chen
- Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Kristina M Chomiak
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, NY, United States of America
| | - Anna Christina Tyler
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, NY, United States of America
| | - Nathan C Eddingsaas
- School of Chemistry and Materials Science, Rochester Institute of Technology, Rochester, NY, United States of America
| | - Sandra E Fischer
- Toronto General Hospital, University Health Network, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Timothy D Jackson
- Division of Surgery, University of Toronto, Toronto, Canada; Division of General Surgery, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Allan Okrainec
- Division of Surgery, University of Toronto, Toronto, Canada; Division of General Surgery, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Johane P Allard
- Toronto General Hospital, University Health Network, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Department of Medicine, Division of Gastroenterology, University of Toronto, Toronto, Canada.
| |
Collapse
|
10
|
Herfindal AM, Nilsen M, Aspholm TE, Schultz GIG, Valeur J, Rudi K, Thoresen M, Lundin KEA, Henriksen C, Bøhn SK. Effects of fructan and gluten on gut microbiota in individuals with self-reported non-celiac gluten/wheat sensitivity-a randomised controlled crossover trial. BMC Med 2024; 22:358. [PMID: 39227818 PMCID: PMC11373345 DOI: 10.1186/s12916-024-03562-1] [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: 05/15/2024] [Accepted: 08/14/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Individuals with non-celiac gluten/wheat sensitivity (NCGWS) experience improvement in gastrointestinal symptoms following a gluten-free diet. Although previous results have indicated that fructo-oligosaccharides (FOS), a type of short-chain fructans, were more likely to induce symptoms than gluten in self-reported NCGWS patients, the underlying mechanisms are unresolved. METHODS Our main objective was therefore to investigate whether FOS-fructans and gluten affect the composition and diversity of the faecal microbiota (16S rRNA gene sequencing), faecal metabolites of microbial fermentation (short-chain fatty acids [SCFA]; gas chromatography with flame ionization detector), and a faecal biomarker of gut inflammation (neutrophil gelatinase-associated lipocalin, also known as lipocalin 2, NGAL/LCN2; ELISA). In the randomised double-blind placebo-controlled crossover study, 59 participants with self-reported NCGWS underwent three different 7-day diet challenges with gluten (5.7 g/day), FOS-fructans (2.1 g/day), and placebo separately (three periods, six challenge sequences). RESULTS The relative abundances of certain bacterial taxa were affected differently by the diet challenges. After the FOS-fructan challenge, Fusicatenibacter increased, while Eubacterium (E.) coprostanoligenes group, Anaerotruncus, and unknown Ruminococcaceae genera decreased. The gluten challenge was primarily characterized by increased abundance of Eubacterium xylanophilum group. However, no differences were found for bacterial diversity (α-diversity), overall bacterial community structure (β-diversity), faecal metabolites (SCFA), or NGAL/LCN2. Furthermore, gastrointestinal symptoms in response to FOS-fructans were generally not linked to substantial shifts in the gut bacterial community. However, the reduction in E. coprostanoligenes group following the FOS-fructan challenge was associated with increased gastrointestinal pain. Finally, correlation analysis revealed that changes in gastrointestinal symptoms following the FOS-fructan and gluten challenges were linked to varying bacterial abundances at baseline. CONCLUSIONS In conclusion, while FOS-fructans induced more gastrointestinal symptoms than gluten in the NCGWS patients, we did not find that substantial shifts in the composition nor function of the faecal microbiota could explain these differences in the current study. However, our results indicate that individual variations in baseline bacterial composition/function may influence the gastrointestinal symptom response to both FOS-fructans and gluten. Additionally, the change in E. coprostanoligenes group, which was associated with increased symptoms, implies that attention should be given to these bacteria in future trials investigating the impact of dietary treatments on gastrointestinal symptoms. TRIAL REGISTRATION Clinicaltrials.gov as NCT02464150.
Collapse
Affiliation(s)
- Anne Mari Herfindal
- Faculty of Chemistry, Biotechnology and Food Sciences, Norwegian University of Life Sciences, P. O. Box 5003, N-1432, Ås, Norway
| | - Morten Nilsen
- Faculty of Chemistry, Biotechnology and Food Sciences, Norwegian University of Life Sciences, P. O. Box 5003, N-1432, Ås, Norway
| | - Trude E Aspholm
- Faculty of Chemistry, Biotechnology and Food Sciences, Norwegian University of Life Sciences, P. O. Box 5003, N-1432, Ås, Norway
| | | | - Jørgen Valeur
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Knut Rudi
- Faculty of Chemistry, Biotechnology and Food Sciences, Norwegian University of Life Sciences, P. O. Box 5003, N-1432, Ås, Norway
| | - Magne Thoresen
- Department of Biostatistics, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Knut E A Lundin
- Disease Research Centre, Norwegian Coeliac, University of Oslo, Oslo, Norway
- Department of Gastroenterology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Christine Henriksen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Siv K Bøhn
- Faculty of Chemistry, Biotechnology and Food Sciences, Norwegian University of Life Sciences, P. O. Box 5003, N-1432, Ås, Norway.
| |
Collapse
|
11
|
Alonso-Allende J, Milagro FI, Aranaz P. Health Effects and Mechanisms of Inulin Action in Human Metabolism. Nutrients 2024; 16:2935. [PMID: 39275251 PMCID: PMC11397174 DOI: 10.3390/nu16172935] [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: 07/29/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 09/16/2024] Open
Abstract
Inulin is a plant polysaccharide which, due to its chemical structure, is not digestible by human gut enzymes but by some bacteria of the human microbiota, acting as a prebiotic. Consequently, inulin consumption has been associated with changes in the composition of the intestinal microbiota related to an improvement of the metabolic state, counteracting different obesity-related disturbances. However, the specific mechanisms of action, including bacterial changes, are not exactly known. Here, a bibliographic review was carried out to study the main effects of inulin on human metabolic health, with a special focus on the mechanisms of action of this prebiotic. Inulin supplementation contributes to body weight and BMI control, reduces blood glucose levels, improves insulin sensitivity, and reduces inflammation markers, mainly through the selective favoring of short-chain fatty acid (SCFA)-producer species from the genera Bifidobacterium and Anaerostipes. These SCFAs have been shown to ameliorate glucose metabolism and decrease hepatic lipogenesis, reduce inflammation, modulate immune activity, and improve anthropometric parameters such as body weight or BMI. In conclusion, the studies collected suggest that inulin intake produces positive metabolic effects through the improvement of the intestinal microbiota and through the metabolites produced by its fermentation.
Collapse
Affiliation(s)
- Jaime Alonso-Allende
- Department of Nutrition, Food Science and Physiology, Faculty of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain
- Center for Nutrition Research, University of Navarra, 31008 Pamplona, Spain
| | - Fermín I Milagro
- Department of Nutrition, Food Science and Physiology, Faculty of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain
- Center for Nutrition Research, University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31009 Pamplona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Paula Aranaz
- Center for Nutrition Research, University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31009 Pamplona, Spain
| |
Collapse
|
12
|
Rahayu ES, Yoga WK, Komalasari H, Mariyatun M, Yuda WA, Manurung NEP, Hasan PN, Suharman S, Pamungkaningtyas FH, Nurfiana DA, Pramesi PC, Gatya M, Therdtatha P, Nakayama J, Juffrie M, Djaafar TF, Marwati T, Utami T. Probiotic Chocolate Containing Lactobacillus plantarum Dad-13 Alters the Gut Microbiota Composition of Undernourished Children in Lombok: A Randomized Double-Blind Trial. INTERNATIONAL JOURNAL OF FOOD SCIENCE 2024; 2024:9493797. [PMID: 39132547 PMCID: PMC11316911 DOI: 10.1155/2024/9493797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 06/11/2024] [Accepted: 07/08/2024] [Indexed: 08/13/2024]
Abstract
The present study investigated the ingestion effect of chocolate probiotic containing Lactobacillus plantarum Dad-13 in undernourished children. A 100-day observation was conducted on undernourished children in Lombok, who were divided into probiotic (n = 28) and placebo (n = 28) groups. Fecal sampling was performed on the 10th and 100th days and further analyzed for gut microbiota composition, short-chain fatty acid (SCFA), and fecal pH. A significant difference was found in the diversity index, fecal pH, and several microbiotas at the phylum and genus levels. At the phylum level, Bacteroidetes was significantly higher in the probiotic group, and a higher relative abundance (RA) of Firmicutes was found in the placebo group. At the genus level, significant differences were observed in some bacteria, such as Bifidobacterium and Prevotella. Therefore, it can be concluded that the probiotic intervention in this study resulted in changes of gut microbiota diversity and fecal pH. Trial Registration: Thai Clinical Trials Registry identifier: TCTR20220425001.
Collapse
Affiliation(s)
- Endang S. Rahayu
- Faculty of Agricultural TechnologyUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
- Center for Food and Nutrition StudiesUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Wahyu K. Yoga
- Faculty of Agricultural TechnologyUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Husnita Komalasari
- Faculty of Agricultural TechnologyUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Mariyatun Mariyatun
- Center for Food and Nutrition StudiesUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | | | - Nancy E. P. Manurung
- Food Technology Study ProgramDepartment of Chemical EngineeringPoliteknik Negeri Sriwijaya, Palembang 30128, Indonesia
| | - Pratama N. Hasan
- Center for Food and Nutrition StudiesUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Suharman Suharman
- Faculty of Agricultural TechnologyUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | | | - Dina A. Nurfiana
- Center for Food and Nutrition StudiesUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Putrika C. Pramesi
- Center for Food and Nutrition StudiesUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Mifta Gatya
- Center for Food and Nutrition StudiesUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | | | - Jiro Nakayama
- Department of Bioscience and BiotechnologyFaculty of AgricultureKyushu University, Fukuoka 819-0395, Japan
| | - Mohammad Juffrie
- Department of Public HealthUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Titiek F. Djaafar
- Research Center for Food Technology and ProcessNational Research and Innovation Agency, Yogyakarta 55861, Indonesia
| | - Tri Marwati
- Research Center for Food Technology and ProcessNational Research and Innovation Agency, Yogyakarta 55861, Indonesia
| | - Tyas Utami
- Faculty of Agricultural TechnologyUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| |
Collapse
|
13
|
Gong T, Liu X, Wang X, Lu Y, Wang X. Applications of polysaccharides in enzyme-triggered oral colon-specific drug delivery systems: A review. Int J Biol Macromol 2024; 275:133623. [PMID: 38969037 DOI: 10.1016/j.ijbiomac.2024.133623] [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: 01/30/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
Enzyme-triggered oral colon-specific drug delivery system (EtOCDDS1) can withstand the harsh stomach and small intestine environments, releasing encapsulated drugs selectively in the colon in response to colonic microflora, exerting local or systematic therapeutic effects. EtOCDDS boasts high colon targetability, enhanced drug bioavailability, and reduced systemic side effects. Polysaccharides are extensively used in enzyme-triggered oral colon-specific drug delivery systems, and its colon targetability has been widely confirmed, as their properties meet the demand of EtOCDDS. Polysaccharides, known for their high safety and excellent biocompatibility, feature modifiable structures. Some remain undigested in the stomach and small intestine, whether in their natural state or after modifications, and are exclusively broken down by colon-resident microbiota. Such characteristics make them ideal materials for EtOCDDS. This article reviews the design principles of EtOCDDS as well as commonly used polysaccharides and their characteristics, modifications, applications and specific mechanism for colon targeting. The article concludes by summarizing the limitations and potential of ETOCDDS to stimulate the development of innovative design approaches.
Collapse
Affiliation(s)
- Tingting Gong
- Institute of Medicinal Plant Development, Peking Union Medical College, No.151, Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Xinxin Liu
- Institute of Medicinal Plant Development, Peking Union Medical College, No.151, Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Xi Wang
- Institute of Medicinal Plant Development, Peking Union Medical College, No.151, Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Yunqian Lu
- Institute of Medicinal Plant Development, Peking Union Medical College, No.151, Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Xiangtao Wang
- Institute of Medicinal Plant Development, Peking Union Medical College, No.151, Malianwa North Road, Haidian District, Beijing 100193, PR China.
| |
Collapse
|
14
|
Reshef N, Gophna U, Reshef L, Konikoff F, Gabay G, Zornitzki T, Knobler H, Maor Y. Prebiotic Treatment in Patients with Nonalcoholic Fatty Liver Disease (NAFLD)-A Randomized Pilot Trial. Nutrients 2024; 16:1571. [PMID: 38892505 PMCID: PMC11174003 DOI: 10.3390/nu16111571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/10/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Several studies show that gut microbiotas in patients with nonalcoholic fatty liver disease (NAFLD) differ from those in a healthy population, suggesting that this alteration plays a role in NAFLD pathogenesis. We investigated whether prebiotic administration affects liver fat content and/or liver-related and metabolic parameters. Patients with NAFLD and metabolic syndrome (age: 50 ± 11; 79% men) were randomized to receive either 16 g/day of prebiotic (ITFs-inulin-type fructans) (n = 8) or placebo (maltodextrin) (n = 11) for 12 weeks. Patients were instructed to maintain a stable weight throughout the study. Liver fat content (measured by H1MRS), fecal microbiota, and metabolic, inflammatory, and liver parameters were determined before and after intervention. Fecal samples from patients who received the prebiotic had an increased content of Bifidobacterium (p = 0.025), which was not observed with the placebo. However, the baseline and end-of-study liver fat contents did not change significantly in the prebiotic and placebo groups, neither did the liver function tests' metabolic and inflammatory mediators, including fibroblast growth factor-19 and lipopolysaccharide-binding protein. Body weight remained stable in both groups. These findings suggest that prebiotic treatment without weight reduction is insufficient to improve NAFLD.
Collapse
Affiliation(s)
- Naama Reshef
- Institute of Diabetes and Metabolism-Kaplan Medical Center, Rehovot 7661041, Israel; (T.Z.); (H.K.)
- School of Nutritional Sciences, Faculty of Agriculture, Food and Environment, The Hebrew University, Jerusalem 9112102, Israel
| | - Uri Gophna
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel-Aviv University, Tel Aviv 6423906, Israel; (U.G.); (L.R.)
| | - Leah Reshef
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel-Aviv University, Tel Aviv 6423906, Israel; (U.G.); (L.R.)
| | - Fred Konikoff
- Institute of Gastroenterology and Hepatology-Meir Medical Center, Kefar Sava 4428164, Israel; (F.K.)
- Faculty of Medicine, Tel-Aviv University, Tel Aviv 6423906, Israel
| | - Gila Gabay
- Institute of Gastroenterology and Hepatology-Meir Medical Center, Kefar Sava 4428164, Israel; (F.K.)
| | - Taiba Zornitzki
- Institute of Diabetes and Metabolism-Kaplan Medical Center, Rehovot 7661041, Israel; (T.Z.); (H.K.)
- Hadassah School of Medicine, The Hebrew University, Jerusalem 9112102, Israel;
| | - Hilla Knobler
- Institute of Diabetes and Metabolism-Kaplan Medical Center, Rehovot 7661041, Israel; (T.Z.); (H.K.)
- Hadassah School of Medicine, The Hebrew University, Jerusalem 9112102, Israel;
| | - Yaakov Maor
- Hadassah School of Medicine, The Hebrew University, Jerusalem 9112102, Israel;
- Institute of Gastroenterology and Hepatology-Kaplan Medical Center, Rehovot 7661043, Israel
| |
Collapse
|
15
|
Li XW, Qiu F, Liu Y, Yang JZ, Chen LJ, Li JH, Liu JL, Hsu C, Chen L, Zeng JH, Xie XL, Wang Q. Inulin alleviates perinatal 2-ethylhexyl diphenyl phosphate (EHDPHP) exposure-induced intestinal toxicity by reshaping the gut microbiota and suppressing the enteric-origin LPS/TLR4/NF-κb pathway in dams and pups. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 346:123659. [PMID: 38417603 DOI: 10.1016/j.envpol.2024.123659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/18/2024] [Accepted: 02/24/2024] [Indexed: 03/01/2024]
Abstract
Organophosphorus flame retardants (OPFRs), such as 2-ethylhexyl diphenyl phosphate (EHDPHP), are ubiquitously used, leading to pervasive environmental contamination and human health risks. While associations between EHDPHP and health issues such as disruption of hormones, neurotoxic effects, and toxicity to reproduction have been recognized, exposure to EHDPHP during perinatal life and its implications for the intestinal health of dams and their pups have largely been unexplored. This study investigated the intestinal toxicity of EHDPHP and the potential for which inulin was effective. Dams were administered either an EHDPHP solution or a corn oil control from gestation day 7 (GD7) to postnatal day 21 (PND21), with inulin provided in their drinking water. Our results indicate that inulin supplementation mitigates damage to the intestinal epithelium caused by EHDPHP, restores mucus-secreting cells, suppresses intestinal hyperpermeability, and abates intestinal inflammation by curtailing lipopolysaccharide leakage through reshaping of the gut microbiota. A reduction in LPS levels concurrently inhibited the inflammation-associated TLR4/NF-κB pathway. In conclusion, inulin administration may ameliorate intestinal toxicity caused by EHDPHP in dams and pups by reshaping the gut microbiota and suppressing the LPS/TLR4/NF-κB pathway. These findings underscore the efficacy of inulin as a therapeutic agent for managing health risks linked to EHDPHP exposure.
Collapse
Affiliation(s)
- Xiu-Wen Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Feng Qiu
- Department of Laboratory Medicine, The Seventh Affiliated Hospital of Southern Medical University, Foshan, Guangdong 528244, China
| | - Yi Liu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jian-Zheng Yang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Li-Jian Chen
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jia-Hao Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jia-Li Liu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Clare Hsu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Long Chen
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jia-Hao Zeng
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Xiao-Li Xie
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), Guangzhou, Guangdong 510515, China
| | - Qi Wang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
16
|
An R, Zhou X, Zhang J, Yang Y, Lyu C, Wang D. Restoration of Intestinal Microbiota After Inulin Supplementation Halted: The Secondary Effect of Supplemented Inulin. Mol Nutr Food Res 2024; 68:e2400033. [PMID: 38483096 DOI: 10.1002/mnfr.202400033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/19/2024] [Indexed: 04/17/2024]
Abstract
SCOPE Consumption of inulin could affect the intestinal microbiota composition. Hereby, it is aimed to investigate the intestinal microbial community restoration process when the inulin supplementation is terminated (i.e., the secondary effect). METHODS AND RESULTS The current study investigates the response and restoration of intestinal microbiota to/after high (Inulin-H) and low (Inulin-L) dosage of inulin supplementation or sequential antibiotics and inulin (Anti-Inulin-L) supplementation, based on analysis of 16S rRNA gene sequences in C57BL/6 mice. The number of significantly changed genera in response to inulin is highest in Anti-Inulin-L (n = 66) group, followed by Inulin-H (n = 51) and Inulin-L (n = 38) group. After inulin supplementation stops, microbiota of all studied groups tend to recover to their original states, with highest percentage of inulin-responding microbes stay significantly different at Anti-Inulin-L (93.94%) group, followed by Inulin-H (74.51%) and Inulin-L (44.12%) groups. Of note, the relative abundance of some non-inulin-responding taxa significantly increases during restoration. CONCLUSION Sequential antibiotics and inulin supplementation induce greatest changes in the intestinal microbial composition, followed by high and low dosage of inulin. Additionally, the changes induce by supplemented inulin in the intestinal microbial community, provide a chance for some microbes to outcompete the other microbes during the spontaneous restoration.
Collapse
Affiliation(s)
- Ran An
- School of Agriculture and Biology, Shanghai Jiao Tong University, Dongchuan 800, Shanghai, 200240, China
| | - Xilong Zhou
- State Key Laboratory of Dairy Biotechnology, Dairy Research Institute, Bright Dairy and Food Co., Ltd, Shanghai, China, Jiangchang West Road 1518, Shanghai, 200436, China
| | - Jing Zhang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Dongchuan 800, Shanghai, 200240, China
| | - Yaqi Yang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Dongchuan 800, Shanghai, 200240, China
| | - Chengang Lyu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Dongchuan 800, Shanghai, 200240, China
| | - Dapeng Wang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Dongchuan 800, Shanghai, 200240, China
| |
Collapse
|
17
|
Ribeiro MC, Levi Y, Moraschini V, Messora MR, Furlaneto FAC. Effects of Prebiotic Therapy on Gastrointestinal Microbiome of Individuals with Different Inflammatory Conditions: A Systematic Review of Randomized Controlled Trials. Probiotics Antimicrob Proteins 2024; 16:673-695. [PMID: 37093515 DOI: 10.1007/s12602-023-10075-5] [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] [Accepted: 04/06/2023] [Indexed: 04/25/2023]
Abstract
Prebiotics are substrates selectively utilized by host microorganisms conferring a health benefit. The effects of prebiotics on the gut microbiome of individuals with inflammatory processes need further investigations. The purpose of this study was to evaluate the effects of prebiotics on the gastrointestinal microbiome of individuals with some types of inflammatory conditions. Randomized controlled clinical trials (RCTs) evaluating the effects of different prebiotics on the gut microbiome were included. A systematic review of the literature including searches in PubMed/MEDLINE, EMBASE, Cochrane Library, Web of Science, and Scopus databases was performed until 23 March 2023. The risk of bias was assessed using the Cochrane Collaboration's criteria. Qualitative data was tabulated to facilitate comparisons and represented in the form of descriptive statistics and summary tables. Thirty trials, ranging from 12 to 135 patients, were included. The most commonly used prebiotic type was inulin-type fructans, and the treatment duration ranged from 1 to 36 weeks. The majority of the trials investigated the gut microbiome using 16 s rRNA gene sequencing on the Illumina Miseq platform. In general, prebiotic therapy exerted positive effects on inflammatory conditions. An increase in Bifidobacterium genus was the most common shift in bacterial composition observed. Within the limits of this systematic review, it can be suggested that prebiotic therapy presents the potential to favorably modulate the gastrointestinal microbiome of individuals with different types of inflammatory conditions.
Collapse
Affiliation(s)
- M C Ribeiro
- Department of Oral Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo - USP, Av. Café S/N, 14020-150, Ribeirao Preto, São Paulo, Brazil
| | - Ylas Levi
- Department of Oral Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo - USP, Av. Café S/N, 14020-150, Ribeirao Preto, São Paulo, Brazil
| | - V Moraschini
- Department of Periodontology, Dental Research Division, School of Dentistry, Veiga de Almeida University, Rio de Janeiro, Brazil
| | - M R Messora
- Department of Oral Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo - USP, Av. Café S/N, 14020-150, Ribeirao Preto, São Paulo, Brazil
| | - F A C Furlaneto
- Department of Oral Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo - USP, Av. Café S/N, 14020-150, Ribeirao Preto, São Paulo, Brazil.
| |
Collapse
|
18
|
Pokushalov E, Ponomarenko A, Garcia C, Pak I, Shrainer E, Seryakova M, Johnson M, Miller R. The Impact of Glucomannan, Inulin, and Psyllium Supplementation (Soloways TM) on Weight Loss in Adults with FTO, LEP, LEPR, and MC4R Polymorphisms: A Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients 2024; 16:557. [PMID: 38398881 PMCID: PMC10892568 DOI: 10.3390/nu16040557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
This study aimed to determine the impact of a fiber supplement on body weight and composition in individuals with obesity with specific genetic polymorphisms. It involved 112 adults with obesity, each with at least one minor allele in the FTO, LEP, LEPR, or MC4R polymorphism. Participants were randomized to receive either a fiber supplement (glucomannan, inulin, and psyllium) or a placebo for 180 days. The experimental group showed significant reductions in body weight (treatment difference: -4.9%; 95% CI: -6.9% to -2.9%; p < 0.01) and BMI (treatment difference: -1.4 kg/m2; 95% CI: -1.7 to -1.2; p < 0.01) compared to placebo. Further significant decreases in fat mass (treatment difference: -13.0%; 95% CI: -14.4 to -11.7; p < 0.01) and visceral fat rating (treatment difference: -1.3; 95% CI: -1.6 to -1.0; p < 0.01) were noted. Homozygous minor allele carriers experienced greater decreases in body weight (treatment difference: -3.2%; 95% CI: -4.9% to -1.6%; p < 0.01) and BMI (treatment difference: -1.2 kg/m2; 95% CI: -2.0 to -0.4; p < 0.01) compared to heterozygous allele carriers. These carriers also had a more significant reduction in fat mass (treatment difference: -9.8%; 95% CI: -10.6 to -9.1; p < 0.01) and visceral fat rating (treatment difference: -0.9; 95% CI: -1.3 to -0.5; p < 0.01). A high incidence of gastrointestinal events was reported in the experimental group (74.6%), unlike the placebo group, which reported no side effects. Dietary supplementation with glucomannan, inulin, and psyllium effectively promotes weight loss and improves body composition in individuals with obesity, particularly those with specific genetic polymorphisms.
Collapse
Affiliation(s)
- Evgeny Pokushalov
- Center for New Medical Technologies, Novosibirsk 630090, Russia; (A.P.); (I.P.); (E.S.); (M.S.)
- Scientific Research Laboratory, Triangel Scientific, San Francisco, CA 94101, USA; (C.G.)
| | - Andrey Ponomarenko
- Center for New Medical Technologies, Novosibirsk 630090, Russia; (A.P.); (I.P.); (E.S.); (M.S.)
| | - Claire Garcia
- Scientific Research Laboratory, Triangel Scientific, San Francisco, CA 94101, USA; (C.G.)
| | - Inessa Pak
- Center for New Medical Technologies, Novosibirsk 630090, Russia; (A.P.); (I.P.); (E.S.); (M.S.)
| | - Evgenya Shrainer
- Center for New Medical Technologies, Novosibirsk 630090, Russia; (A.P.); (I.P.); (E.S.); (M.S.)
| | - Mariya Seryakova
- Center for New Medical Technologies, Novosibirsk 630090, Russia; (A.P.); (I.P.); (E.S.); (M.S.)
| | - Michael Johnson
- Scientific Research Laboratory, Triangel Scientific, San Francisco, CA 94101, USA; (C.G.)
| | - Richard Miller
- Scientific Research Laboratory, Triangel Scientific, San Francisco, CA 94101, USA; (C.G.)
| |
Collapse
|
19
|
Shtossel O, Koren O, Shai I, Rinott E, Louzoun Y. Gut microbiome-metabolome interactions predict host condition. MICROBIOME 2024; 12:24. [PMID: 38336867 PMCID: PMC10858481 DOI: 10.1186/s40168-023-01737-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 12/10/2023] [Indexed: 02/12/2024]
Abstract
BACKGROUND The effect of microbes on their human host is often mediated through changes in metabolite concentrations. As such, multiple tools have been proposed to predict metabolite concentrations from microbial taxa frequencies. Such tools typically fail to capture the dependence of the microbiome-metabolite relation on the environment. RESULTS We propose to treat the microbiome-metabolome relation as the equilibrium of a complex interaction and to relate the host condition to a latent representation of the interaction between the log concentration of the metabolome and the log frequencies of the microbiome. We develop LOCATE (Latent variables Of miCrobiome And meTabolites rElations), a machine learning tool to predict the metabolite concentration from the microbiome composition and produce a latent representation of the interaction. This representation is then used to predict the host condition. LOCATE's accuracy in predicting the metabolome is higher than all current predictors. The metabolite concentration prediction accuracy significantly decreases cross datasets, and cross conditions, especially in 16S data. LOCATE's latent representation predicts the host condition better than either the microbiome or the metabolome. This representation is strongly correlated with host demographics. A significant improvement in accuracy (0.793 vs. 0.724 average accuracy) is obtained even with a small number of metabolite samples ([Formula: see text]). CONCLUSION These results suggest that a latent representation of the microbiome-metabolome interaction leads to a better association with the host condition than any of the two separated or the simple combination of the two. Video Abstract.
Collapse
Affiliation(s)
- Oshrit Shtossel
- Department of Mathematics, Bar-Ilan University, Ramat Gan, 52900, Israel
| | - Omry Koren
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Iris Shai
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ehud Rinott
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yoram Louzoun
- Department of Mathematics, Bar-Ilan University, Ramat Gan, 52900, Israel.
| |
Collapse
|
20
|
Talukdar JR, Cooper M, Lyutvyn L, Zeraatkar D, Ali R, Berbrier R, Janes S, Ha V, Darling PB, Xue M, Chu A, Chowdhury F, Harnack HE, Huang L, Malik M, Powless J, Lavergne FV, Zhang X, Ehrlich S, Jenkins DJ, Sievenpiper JL, Banfield L, Mbuagbaw L, de Souza RJ. The effects of inulin-type fructans on cardiovascular disease risk factors: systematic review and meta-analysis of randomized controlled trials. Am J Clin Nutr 2024; 119:496-510. [PMID: 38309832 DOI: 10.1016/j.ajcnut.2023.10.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/21/2023] [Accepted: 10/24/2023] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND Inulin-type fructans (ITF) are the leading prebiotics in the market. Available evidence provides conflicting results regarding the beneficial effects of ITF on cardiovascular disease risk factors. OBJECTIVES This study aimed to evaluate the effects of ITF supplementation on cardiovascular disease risk factors in adults. METHODS We searched MEDLINE, EMBASE, Emcare, AMED, CINAHL, and the Cochrane Library databases from inception through May 15, 2022. Eligible randomized controlled trials (RCTs) administered ITF or placebo (for example, control, foods, diets) to adults for ≥2 weeks and reported one or more of the following: low, very-low, or high-density lipoprotein cholesterol (LDL-C, VLDL-C, HDL-C); total cholesterol; apolipoprotein A1 or B; triglycerides; fasting blood glucose; body mass index; body weight; waist circumference; waist-to-hip ratio; systolic or diastolic blood pressure; or hemoglobin A1c. Two reviewers independently and in duplicate screened studies, extracted data, and assessed risk of bias. We pooled data using random-effects model, and assessed the certainty of evidence (CoE) using the Grading of Recommendations, Assessment, Development and Evaluation approach. RESULTS We identified 1767 studies and included 55 RCTs with 2518 participants in meta-analyses. The pooled estimate showed that ITF supplementation reduced LDL-C [mean difference (MD) -0.14 mmol/L, 95% confidence interval (95% CI: -0.24, -0.05), 38 RCTs, 1879 participants, very low CoE], triglycerides (MD -0.06 mmol/L, 95% CI: -0.12, -0.01, 40 RCTs, 1732 participants, low CoE), and body weight (MD -0.97 kg, 95% CI: -1.28, -0.66, 36 RCTs, 1672 participants, low CoE) but little to no significant effect on other cardiovascular disease risk factors. The effects were larger when study duration was ≥6 weeks and in pre-obese and obese participants. CONCLUSION ITF may reduce low-density lipoprotein, triglycerides, and body weight. However, due to low to very low CoE, further well-designed and executed trials are needed to confirm these effects. PROSPERO REGISTRATION NUMBER CRD42019136745.
Collapse
Affiliation(s)
- Jhalok Ronjan Talukdar
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Matthew Cooper
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Lyuba Lyutvyn
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Dena Zeraatkar
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada; Blavatnik Institute, Harvard Medical School, Boston, MA, United States
| | - Rahim Ali
- Faculty of Law, Common Law Section, University of Ottawa, Ottawa, ON, Canada
| | - Rachel Berbrier
- Division of Dermatology, McGill University Health Centre, Montreal, QC, Canada
| | - Sabrina Janes
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Vanessa Ha
- School of Medicine, Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | - Pauline B Darling
- School of Nutrition Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Mike Xue
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Alexandro Chu
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Fariha Chowdhury
- Department of Rehabilitation Science(s), McMaster University, Hamilton, ON, Canada
| | - Hope E Harnack
- Department of Kinesiology, Faculty of Science, McMaster University, Hamilton, ON, Canada
| | - Louise Huang
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Mikail Malik
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Jacqui Powless
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Florence V Lavergne
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, ON, Canada
| | - Xuehong Zhang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Shelley Ehrlich
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - David Ja Jenkins
- Clinical Nutrition and Risk Factor Modification Centre, St. Michael's Hospital, Toronto, ON, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada; Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Toronto 3D Knowledge Synthesis and Clinical Trials Unit, St. Michael's Hospital, Toronto, ON, Canada; Division of Endocrinology and Metabolism, Department of Medicine, St. Michael's Hospital, Toronto, ON, Canada
| | - John L Sievenpiper
- Clinical Nutrition and Risk Factor Modification Centre, St. Michael's Hospital, Toronto, ON, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada; Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Toronto 3D Knowledge Synthesis and Clinical Trials Unit, St. Michael's Hospital, Toronto, ON, Canada; Division of Endocrinology and Metabolism, Department of Medicine, St. Michael's Hospital, Toronto, ON, Canada
| | - Laura Banfield
- Health Sciences Library, McMaster University, Hamilton, ON, Canada; Global Health Graduate Program, McMaster University, Hamilton, ON, Canada
| | - Lawrence Mbuagbaw
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada; Department of Anesthesia, McMaster University, Hamilton, ON, Canada; Department of Pediatrics, McMaster University, Hamilton, ON, Canada; Biostatistics Unit, Father Sean O'Sullivan Research Centre, St Joseph's Healthcare, Hamilton, ON, Canada; Centre for Development of Best Practices in Health (CDBPH), Yaoundé Central Hospital, Yaoundé, Cameroon; Department of Global Health, Stellenbosch University, Cape Town, South Africa
| | - Russell J de Souza
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada; Toronto 3D Knowledge Synthesis and Clinical Trials Unit, St. Michael's Hospital, Toronto, ON, Canada; Global Health Graduate Program, McMaster University, Hamilton, ON, Canada; Department of Pediatrics, McMaster University, Hamilton, ON, Canada; Population Health Research Institute, Hamilton Health Sciences Corporation, Hamilton, ON, Canada.
| |
Collapse
|
21
|
Akram W, Pandey V, Sharma R, Joshi R, Mishra N, Garud N, Haider T. Inulin: Unveiling its potential as a multifaceted biopolymer in prebiotics, drug delivery, and therapeutics. Int J Biol Macromol 2024; 259:129131. [PMID: 38181920 DOI: 10.1016/j.ijbiomac.2023.129131] [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: 10/22/2023] [Revised: 12/18/2023] [Accepted: 12/27/2023] [Indexed: 01/07/2024]
Abstract
In recent years, inulin has gained much attention as a promising multifunctional natural biopolymer with numerous applications in drug delivery, prebiotics, and therapeutics. It reveals a multifaceted biopolymer with transformative implications by elucidating the intricate interplay between inulin and the host, microbiome, and therapeutic agents. Their flexible structure, exceptional targetability, biocompatibility, inherent ability to control release behavior, tunable degradation kinetics, and protective ability make them outstanding carriers in healthcare and biomedicine. USFDA has approved Inulin as a nutritional dietary supplement for infants. The possible applications of inulin in biomedicine research inspired by nature are presented. The therapeutic potential of inulin goes beyond its role in prebiotics and drug delivery. Recently, significant research efforts have been made towards inulin's anti-inflammatory, antioxidant, and immunomodulatory properties for their potential applications in treating various chronic diseases. Moreover, its ability to reduce inflammation and modulate immune responses opens new avenues for treating conditions such as autoimmune disorders and gastrointestinal ailments. This review will attempt to illustrate the inulin's numerous and interconnected roles, shedding light on its critical contributions to the advancement of healthcare and biomedicine and its recent advancement in therapeutics, and conclude by taking valuable insights into the prospects and opportunities of inulin.
Collapse
Affiliation(s)
- Wasim Akram
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Gwalior, Madhya Pradesh 4774005, India
| | - Vikas Pandey
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Gwalior, Madhya Pradesh 4774005, India
| | - Rajeev Sharma
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Gwalior, Madhya Pradesh 4774005, India
| | - Ramakant Joshi
- Department of Pharmaceutics, ShriRam college of Pharmacy, Banmore 476444, India
| | - Neeraj Mishra
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Gwalior, Madhya Pradesh 4774005, India
| | - Navneet Garud
- School of Studies in Pharmaceutical Sciences, Jiwaji University, Gwalior 474011, India
| | - Tanweer Haider
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Gwalior, Madhya Pradesh 4774005, India.
| |
Collapse
|
22
|
Medawar E, Beyer F, Thieleking R, Haange SB, Rolle-Kampczyk U, Reinicke M, Chakaroun R, von Bergen M, Stumvoll M, Villringer A, Witte AV. Prebiotic diet changes neural correlates of food decision-making in overweight adults: a randomised controlled within-subject cross-over trial. Gut 2024; 73:298-310. [PMID: 37793780 PMCID: PMC10850731 DOI: 10.1136/gutjnl-2023-330365] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/12/2023] [Indexed: 10/06/2023]
Abstract
OBJECTIVE Animal studies suggest that prebiotic, plant-derived nutrients could improve homoeostatic and hedonic brain functions through improvements in microbiome-gut-brain communication. However, little is known if these results are applicable to humans. Therefore, we tested the effects of high-dosed prebiotic fibre on reward-related food decision-making in a randomised controlled within-subject cross-over study and assayed potential microbial and metabolic markers. DESIGN 59 overweight young adults (19 females, 18-42 years, body mass index 25-30 kg/m2) underwent functional task MRI before and after 14 days of supplementary intake of 30 g/day of inulin (prebiotics) and equicaloric placebo, respectively. Short chain fatty acids (SCFA), gastrointestinal hormones, glucose/lipid and inflammatory markers were assayed in fasting blood. Gut microbiota and SCFA were measured in stool. RESULTS Compared with placebo, participants showed decreased brain activation towards high-caloric wanted food stimuli in the ventral tegmental area and right orbitofrontal cortex after prebiotics (preregistered, family wise error-corrected p <0.05). While fasting blood levels remained largely unchanged, 16S-rRNA sequencing showed significant shifts in the microbiome towards increased occurrence of, among others, SCFA-producing Bifidobacteriaceae, and changes in >60 predicted functional signalling pathways after prebiotic intake. Changes in brain activation correlated with changes in Actinobacteria microbial abundance and associated activity previously linked with SCFA production, such as ABC transporter metabolism. CONCLUSIONS In this proof-of-concept study, a prebiotic intervention attenuated reward-related brain activation during food decision-making, paralleled by shifts in gut microbiota. TRIAL REGISTRATION NUMBER NCT03829189.
Collapse
Affiliation(s)
- Evelyn Medawar
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Berlin School of Mind and Brain, Humboldt-Universität zu Berlin, Berlin, Germany
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Frauke Beyer
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
| | - Ronja Thieleking
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Sven-Bastiaan Haange
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Ulrike Rolle-Kampczyk
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Madlen Reinicke
- Institute for Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Rima Chakaroun
- Department of Molecular and Clinical Medicine, University of Gothenburg, Goteborg, Sweden
- Medical Department III Endocrinology Nephrology Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Martin von Bergen
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Michael Stumvoll
- Medical Department III Endocrinology Nephrology Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Arno Villringer
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
| | - A Veronica Witte
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
| |
Collapse
|
23
|
Zhang Q, Xiao X, Zheng J, Li M, Yu M, Ping F, Wang T. Maternal inulin alleviates high-fat diet-induced lipid disorder in offspring by epigenetically modulating hypothalamus feeding circuit-related genes. Food Funct 2024; 15:110-124. [PMID: 38044717 DOI: 10.1039/d3fo02223d] [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: 12/05/2023]
Abstract
Increasing evidence supports the existence of fetal-originated adult diseases. Recent research indicates that the intrauterine environment affects the fetal hypothalamic energy intake center. Inulin is a probiotic that can moderate metabolic disorders, but whether maternal inulin intervention confers long-term metabolic benefits to lipid metabolism in offspring in their adult lives and the mechanism involved are unknown. Here, we used a maternal overnutrition model that was induced by excess energy intake before and during pregnancy and lactation and maternal inulin intervention was performed during pregnancy and lactation. The hypothalamic genome methylation in offspring was analyzed using a methylation array. The results showed that maternal inulin treatment modified the maternal high-fat diet (HFD)-induced increases in body weight, adipose tissue weight, and serum insulin and leptin levels and decreases in serum adiponectin levels. Maternal inulin intervention regulated the impairments in hypothalamic leptin resistance, induced the methylation of Socs3, Npy, and Il6, and inhibited the methylation of Lepr in the hypothalamus of offspring. In conclusion, maternal inulin intervention modifies offspring lipid metabolism, and the underlying mechanism involves the methylation of genes in the hypothalamus feeding circuit.
Collapse
Affiliation(s)
- Qian Zhang
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Xinhua Xiao
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Jia Zheng
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Ming Li
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Miao Yu
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Fan Ping
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Tong Wang
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
24
|
Horvath A, Zukauskaite K, Hazia O, Balazs I, Stadlbauer V. Human gut microbiome: Therapeutic opportunities for metabolic syndrome-Hype or hope? Endocrinol Diabetes Metab 2024; 7:e436. [PMID: 37771199 PMCID: PMC10781898 DOI: 10.1002/edm2.436] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 06/01/2023] [Accepted: 06/11/2023] [Indexed: 09/30/2023] Open
Abstract
Shifts in gut microbiome composition and metabolic disorders are associated with one another. Clinical studies and experimental data suggest a causal relationship, making the gut microbiome an attractive therapeutic goal. Diet, intake of probiotics or prebiotics and faecal microbiome transplantation (FMT) are methods to alter a person's microbiome composition. Although FMT may allow establishing a proof of concept to use microbiome modulation to treat metabolic disorders, studies show mixed results regarding the effects on metabolic parameters as well as on the composition of the microbiome. This review summarizes the current knowledge on diet, probiotics, prebiotics and FMT to treat metabolic diseases, focusing on studies that also report alterations in microbiome composition. Furthermore, clinical trial results on the effects of common drugs used to treat metabolic diseases are synopsized to highlight the bidirectional relationship between the microbiome and metabolic diseases. In conclusion, there is clear evidence that microbiome modulation has the potential to influence metabolic diseases; however, it is not possible to distinguish which intervention is the most successful. In addition, a clear commitment from all stakeholders is necessary to move forward in the direction of developing targeted interventions for microbiome modulation.
Collapse
Affiliation(s)
- Angela Horvath
- Medical University of GrazGrazAustria
- Center for Biomarker Research in Medicine (CBmed)GrazAustria
| | - Kristina Zukauskaite
- Medical University of GrazGrazAustria
- Life Sciences CentreVilnius UniversityVilniusLithuania
| | - Olha Hazia
- Medical University of GrazGrazAustria
- Center for Biomarker Research in Medicine (CBmed)GrazAustria
| | - Irina Balazs
- Medical University of GrazGrazAustria
- Center for Biomarker Research in Medicine (CBmed)GrazAustria
| | - Vanessa Stadlbauer
- Medical University of GrazGrazAustria
- Center for Biomarker Research in Medicine (CBmed)GrazAustria
| |
Collapse
|
25
|
Ribeiro PVDM, Veloso TG, de Oliveira LL, Mendes NP, Alfenas RDCG. Consumption of yacon flour and energy-restricted diet increased the relative abundance of intestinal bacteria in obese adults. Braz J Microbiol 2023; 54:3085-3099. [PMID: 37807018 PMCID: PMC10689717 DOI: 10.1007/s42770-023-01140-w] [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: 04/13/2023] [Accepted: 09/26/2023] [Indexed: 10/10/2023] Open
Abstract
Prebiotics can alter the gastrointestinal environment, favoring the growth of health-promoting bacteria. Although yacon is a functional food, with prebiotic properties (fructooligosaccharides), its effects on the intestinal microbiota have not been investigated yet. The objective of this study was to evaluate the effects of yacon flour consumption and energy-restricted diet in the intestinal microbiota in adults with excess body weight. Twenty-one adults with excess body weight were included in this randomized, parallel, double-blind, placebo-controlled, 6-week clinical trial. Subjects daily consumed at breakfast a drink containing 25 g of yacon flour (n = 11) or not containing yacon (n = 10) and received the prescription of energy-restricted diets. Fecal samples were collected on the first and on last day of the study. 16S rRNA sequencing was assessed to evaluate the effect of yacon fermentation on intestinal microbiota bacterial composition. There was an increase in the genera Bifidobacterium, Blautia, Subdoligranulum, and Streptococcus after the consumption of yacon and energy-restricted diet. In the yacon group, we also observed a positive correlation between the concentrations of short-chain fatty acids versus the genera Coprococcus and Howardella, besides a negative correlation between the concentrations of advanced glycation end products and early glycation products versus the genera Ruminococcus and Prevotella, respectively. Consumption of yacon flour and energy-restricted diet selectively changed the intestinal microbiota composition in adults with excess body weight. TRIAL REGISTRATION: Register number: RBR-6YH6BQ. Registered 23 January, 2018.
Collapse
Affiliation(s)
- Priscila Vaz de Melo Ribeiro
- Department of Nutrition and Health, Universidade Federal de Viçosa, Avenida Peter Henry Rolfs, s/n, Campus Universitário, Viçosa, Minas Gerais, 36570-900, Brazil
| | - Tomas Gomes Veloso
- Department of Microbiology, Universidade Federal de Viçosa, Avenida Peter Henry Rolfs, s/n, Campus Universitário, Viçosa, Minas Gerais, 36570-900, Brazil
| | - Leandro Licursi de Oliveira
- Department of General Biology, Universidade Federal de Viçosa, Avenida Peter Henry Rolfs, s/n, Campus Universitário, Viçosa, Minas Gerais, 36570-900, Brazil
| | - Nélia Pinheiro Mendes
- Department of Nutrition and Health, Universidade Federal de Viçosa, Avenida Peter Henry Rolfs, s/n, Campus Universitário, Viçosa, Minas Gerais, 36570-900, Brazil.
| | - Rita de Cássia Gonçalves Alfenas
- Department of Nutrition and Health, Universidade Federal de Viçosa, Avenida Peter Henry Rolfs, s/n, Campus Universitário, Viçosa, Minas Gerais, 36570-900, Brazil
| |
Collapse
|
26
|
Sababathy M, Ramanathan G, Abd Rahaman NY, Ramasamy R, Biau FJ, Qi Hao DL, Hamid NFS. A 'one stone, two birds' approach with mesenchymal stem cells for acute respiratory distress syndrome and Type II diabetes mellitus. Regen Med 2023; 18:913-934. [PMID: 38111999 DOI: 10.2217/rme-2023-0193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023] Open
Abstract
This review explores the intricate relationship between acute respiratory distress syndrome (ARDS) and Type II diabetes mellitus (T2DM). It covers ARDS epidemiology, etiology and pathophysiology, along with current treatment trends and challenges. The lipopolysaccharides (LPS) role in ARDS and its association between non-communicable diseases and COVID-19 are discussed. The review highlights the therapeutic potential of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) for ARDS and T2DM, emphasizing their immunomodulatory effects. This review also underlines how T2DM exacerbates ARDS pathophysiology and discusses the potential of hUC-MSCs in modulating immune responses. In conclusion, the review highlights the multidisciplinary approach to managing ARDS and T2DM, focusing on inflammation, oxidative stress and potential therapy of hUC-MSCs in the future.
Collapse
Affiliation(s)
- Mogesh Sababathy
- Department of Veterinary Pathology & Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Ghayathri Ramanathan
- Faculty of Computer Science & Information Technology, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Nor Yasmin Abd Rahaman
- Department of Veterinary Laboratory Diagnostics, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Laboratory of Vaccines & Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Rajesh Ramasamy
- Department of Pathology, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Foo Jhi Biau
- Centre for Drug Discovery & Molecular Pharmacology (CDDMP), Faculty of Health & Medical Sciences, Taylor's University, Selangor, Subang Jaya, 47500, Malaysia
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, Selangor, Subang Jaya, 47500, Malaysia
| | - Daniel Looi Qi Hao
- My Cytohealth Sdn. Bhd., 18-2, Jalan Radin Bagus 1, Bandar Seri Petaling, Kuala Lumpur, 57000, Malaysia
| | - Nur-Fazila Saulol Hamid
- Department of Veterinary Pathology & Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Laboratory of Vaccines & Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| |
Collapse
|
27
|
Ahmad J, Khan I, Zengin G, Mahomoodally MF. The gut microbiome in the fight against obesity: The potential of dietary factors. FASEB J 2023; 37:e23258. [PMID: 37843880 DOI: 10.1096/fj.202300864rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/17/2023]
Abstract
Obesity as a global public health burden has experienced a drastic growing trend recently. The management of obesity is challenging because of its complex etiology, and various factors are involved in its development, such as genetic and environmental factors. Different approaches are available to treat and/or manage obesity, including diet, physical activity, lifestyle changes, medications, and surgery. However, some of these approaches have inherent limitations and are closely associated with adverse effects. Therefore, probing into a novel/safe approach to treat and/or manage obesity is of fundamental importance. One such approach gaining renewed interest is the potential role of gut microbiota in obesity and its effectiveness in treating this condition. However, there is a dearth of comprehensive compilation of data on the potential role of the gut microbiome in obesity, particularly regarding dietary factors as a therapeutic approach. Therefore, this review aims to provide an updated overview of the role of gut microbiota in obesity, further highlighting the importance of dietary factors, particularly diet, prebiotics, and probiotics, as potential complementary and/or alternative therapeutic options. Moreover, the association of gut microbiota with obese or lean individuals has also been discussed.
Collapse
Affiliation(s)
- Jamil Ahmad
- Department of Human Nutrition, The University of Agriculture Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Imran Khan
- Department of Human Nutrition, The University of Agriculture Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, Konya, Turkey
| | - Mohamad Fawzi Mahomoodally
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
- School of Engineering & Technology, Duy Tan University, Da Nang, Vietnam
| |
Collapse
|
28
|
Nagy DU, Sándor-Bajusz KA, Bódy B, Decsi T, Van Harsselaar J, Theis S, Lohner S. Effect of chicory-derived inulin-type fructans on abundance of Bifidobacterium and on bowel function: a systematic review with meta-analyses. Crit Rev Food Sci Nutr 2023; 63:12018-12035. [PMID: 35833477 DOI: 10.1080/10408398.2022.2098246] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Inulin-type fructans are considered to stimulate the growth of beneficial microorganisms, like Bifidobacterium in the gut and support health. However, both the fructan source and chemical structure may modify these effects. A systematic review was conducted to assess the effects of chicory-derived inulin-type fructans consumed either in specific foods or as dietary supplements on abundance of Bifidobacterium in the gut and on health-related outcomes. Three electronic databases and two clinical trial registries were systematically searched until January 2021. Two authors independently selected randomized controlled trials that investigated with a protocol of minimum seven days supplementation the effect of chicory-derived inulin-type fructans on Bifidobacterium abundance in any population. Meta-analyses with random-effects model were conducted on Bifidobacterium abundance and bowel function parameters. We evaluated risk of bias using Cochrane RoB tool. Chicory-derived inulin-type fructans at a dose of 3-20 g/day significantly increased Bifidobacterium abundance in participants with an age range from 0 to 83 years (standardized mean difference: 0.83, 95% CI: 0.58-1.08; p < 0.01; 50 studies; 2525 participants). Significant bifidogenic effects were observed in healthy individuals and in populations with health impairments, except gastrointestinal disorders. Significant beneficial effects on bowel function parameters were observed in healthy subjects. Chicory-derived inulin-type fructans may have significant bifidogenic effects and may beneficially influence bowel function in healthy individuals. PROSPERO registration number CRD42020162892.
Collapse
Affiliation(s)
- Dávid U Nagy
- Department of Paediatrics, Clinical Center of the University of Pécs, Medical School, University of Pécs, Pécs, Hungary
- Institute of Geobotany/Plant Ecology, Martin-Luther-University, Halle (Saale), Germany
| | - Kinga Amália Sándor-Bajusz
- Department of Paediatrics, Clinical Center of the University of Pécs, Medical School, University of Pécs, Pécs, Hungary
| | - Blanka Bódy
- Department of Paediatrics, Clinical Center of the University of Pécs, Medical School, University of Pécs, Pécs, Hungary
| | - Tamás Decsi
- Department of Paediatrics, Clinical Center of the University of Pécs, Medical School, University of Pécs, Pécs, Hungary
| | | | - Stephan Theis
- BENEO-Institute, c/o BENEO GmbH, Obrigheim, (Germany)
| | - Szimonetta Lohner
- Department of Public Health Medicine, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
29
|
Barouei J, Martinic A, Bendiks Z, Mishchuk D, Heeney D, Slupsky CM, Marco ML. Type 2-resistant starch and Lactiplantibacillus plantarum NCIMB 8826 result in additive and interactive effects in diet-induced obese mice. Nutr Res 2023; 118:12-28. [PMID: 37536013 DOI: 10.1016/j.nutres.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 08/05/2023]
Abstract
Little is known about how combining a probiotic with prebiotic dietary fiber affects the ability of either biotic to improve health. We hypothesized that prebiotic, high-amylose maize type 2-resistant starch (RS) together with probiotic Lactiplantibacillus plantarum NCIMB8826 (LP) as a complementary synbiotic results in additive effects on the gut microbiota in diet-induced obese mice and other body sites. Diet-induced obese C57BL/6J male mice were fed a high-fat diet adjusted to contain RS (20% by weight), LP (109 cells every 48 hours), or both (RS+LP) for 6 weeks. As found for mice fed RS, cecal bacterial alpha diversity was significantly reduced in mice given RS+LP compared with those fed LP and high-fat controls. Similarly, both RS+LP and RS also conferred lower quantities of cecal butyrate and serum histidine and higher ileal TLR2 transcript levels and adipose tissue interleukin-6 protein. As found for mice fed LP, RS+LP-fed mice had higher colonic tissue TH17 cytokines, reduced epididymal fat immune and oxidative stress responses, reduced serum carnitine levels, and increased transcript quantities of hepatic carnitine palmitoyl transferase 1α. Notably, compared with RS and LP consumed separately, there were also synergistic increases in colonic glucose and hepatic amino acids as well antagonistic effects of LP on RS-mediated increases in serum adiponectin and urinary toxin levels. Our findings show that it is not possible to fully predict outcomes of synbiotic applications based on findings of the probiotic or the prebiotic tested separately; therefore, studies should be conducted to test new synbiotic formulations.
Collapse
Affiliation(s)
- Javad Barouei
- Integrated Food Security Research Center, College of Agriculture and Human Sciences, Prairie View A&M University, Prairie View, TX; Department of Food Science & Technology, University of California, Davis, CA
| | - Alice Martinic
- Department of Nutrition, University of California, Davis, CA
| | - Zach Bendiks
- Department of Food Science & Technology, University of California, Davis, CA
| | - Darya Mishchuk
- Department of Food Science & Technology, University of California, Davis, CA
| | - Dustin Heeney
- Department of Food Science & Technology, University of California, Davis, CA
| | - Carolyn M Slupsky
- Department of Food Science & Technology, University of California, Davis, CA; Department of Nutrition, University of California, Davis, CA
| | - Maria L Marco
- Department of Food Science & Technology, University of California, Davis, CA.
| |
Collapse
|
30
|
Coe GL, Krout IN, Munro-Ehrlich M, Beamish CR, Vorojeikina D, Colman DR, Boyd EJ, Walk ST, Rand MD. Assessing the role of the gut microbiome in methylmercury demethylation and elimination in humans and gnotobiotic mice. Arch Toxicol 2023; 97:2399-2418. [PMID: 37392210 PMCID: PMC10913183 DOI: 10.1007/s00204-023-03548-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/14/2023] [Indexed: 07/03/2023]
Abstract
The risk of methylmercury (MeHg) toxicity following ingestion of contaminated foodstuffs (e.g., fish) is directly related to the kinetics of MeHg elimination among individuals. Yet, the factors driving the wide range of inter-individual variability in MeHg elimination within a population are poorly understood. Here, we investigated the relationship between MeHg elimination, gut microbiome demethylation activity, and gut microbiome composition using a coordinated human clinical trial and gnotobiotic mouse modeling approach together with metagenomic sequence analysis. We first observed MeHg elimination half-lives (t1/2) ranging from 28 to 90 days across 27 volunteers. Subsequently, we found that ingestion of a prebiotic induced changes in the gut microbiome and mixed effects (increased, decrease, and no effect) on elimination in these same individuals. Nonetheless, elimination rates were found to correlate with MeHg demethylation activity in cultured stool samples. In mice, attempts to remove the microbiome via generation of germ-free (GF) animals or through antibiotic (Abx) treatment both diminished MeHg demethylation to a similar extent. While both conditions substantially slowed elimination, Abx treatment resulted in significantly slower elimination than the GF condition, indicating an additional role for host-derived factors in supporting elimination. Human fecal microbiomes transplanted to GF mice restored elimination rates to that seen in control mice. Metagenomic sequence analysis of human fecal DNA did not identify genes encoding proteins typically involved in demethylation (e.g., merB, organomercury lyase). However, the abundance of several anaerobic taxa, notably Alistipes onderdonkii, were positively correlated with MeHg elimination. Surprisingly, mono-colonization of GF free mice with A. onderdonkii did not restore MeHg elimination to control levels. Collectively, our findings indicate the human gut microbiome uses a non-conventional pathway of demethylation to increase MeHg elimination that relies on yet to be resolved functions encoded by the gut microbes and the hostClinical Trial NCT04060212, prospectively registered 10/1/2019.
Collapse
Affiliation(s)
- Genevieve L Coe
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Ian N Krout
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Mason Munro-Ehrlich
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Catherine R Beamish
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Daria Vorojeikina
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Daniel R Colman
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Eric J Boyd
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Seth T Walk
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Matthew D Rand
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
31
|
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]
|
32
|
González A, Conceição E, Teixeira JA, Nobre C. In vitro models as a tool to study the role of gut microbiota in obesity. Crit Rev Food Sci Nutr 2023; 64:10912-10923. [PMID: 37403775 DOI: 10.1080/10408398.2023.2232022] [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: 07/06/2023]
Abstract
Obesity, a highly prevalent condition worldwide that leads to the development of multiple metabolic diseases, has been related to gut microbial dysbiosis. To understand this correlation, in vivo models have been extremely useful. However, its use is limited by associated ethical concerns, high costs, low representativeness, and low reproducibility. Therefore, new and improved in vitro models have been developed in recent years, representing a promising tool in the study of the role of gut microbiota modulation in weight management and metabolic health. This review aims to provide an update on the main findings obtained in vitro regarding gut microbiota modulation with probiotics, and food compounds, and its interaction with the host metabolism, associated with obesity. Available in vitro colon models currently used to study obesity are discussed, including batch and dynamic fermentation systems, and models that allow the study of microbiota-host interactions using cell cultures. In vitro models have demonstrated that homeostatic microbiota may help overcome obesity by producing satiety-related neurotransmitters and metabolites that protect the gut barrier and improve the metabolic activity of adipose tissue. In vitro models may be the key to finding new treatments for obesity-related disorders.
Collapse
Affiliation(s)
- Abigail González
- CEB - Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Eva Conceição
- CIPsi - Psychology Research Centre, University of Minho Campus de Gualtar, Braga, Portugal
| | - José António Teixeira
- CEB - Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS -Associate Laboratory, Braga, Portugal
| | - Clarisse Nobre
- CEB - Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS -Associate Laboratory, Braga, Portugal
| |
Collapse
|
33
|
Mendoza-León MJ, Mangalam AK, Regaldiz A, González-Madrid E, Rangel-Ramírez MA, Álvarez-Mardonez O, Vallejos OP, Méndez C, Bueno SM, Melo-González F, Duarte Y, Opazo MC, Kalergis AM, Riedel CA. Gut microbiota short-chain fatty acids and their impact on the host thyroid function and diseases. Front Endocrinol (Lausanne) 2023; 14:1192216. [PMID: 37455925 PMCID: PMC10349397 DOI: 10.3389/fendo.2023.1192216] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/23/2023] [Indexed: 07/18/2023] Open
Abstract
Thyroid disorders are clinically characterized by alterations of L-3,5,3',5'-tetraiodothyronine (T4), L-3,5,3'-triiodothyronine (T3), and/or thyroid-stimulating hormone (TSH) levels in the blood. The most frequent thyroid disorders are hypothyroidism, hyperthyroidism, and hypothyroxinemia. These conditions affect cell differentiation, function, and metabolism. It has been reported that 40% of the world's population suffers from some type of thyroid disorder and that several factors increase susceptibility to these diseases. Among them are iodine intake, environmental contamination, smoking, certain drugs, and genetic factors. Recently, the intestinal microbiota, composed of more than trillions of microbes, has emerged as a critical player in human health, and dysbiosis has been linked to thyroid diseases. The intestinal microbiota can affect host physiology by producing metabolites derived from dietary fiber, such as short-chain fatty acids (SCFAs). SCFAs have local actions in the intestine and can affect the central nervous system and immune system. Modulation of SCFAs-producing bacteria has also been connected to metabolic diseases, such as obesity and diabetes. In this review, we discuss how alterations in the production of SCFAs due to dysbiosis in patients could be related to thyroid disorders. The studies reviewed here may be of significant interest to endocrinology researchers and medical practitioners.
Collapse
Affiliation(s)
- María José Mendoza-León
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | | | - Alejandro Regaldiz
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Medicina Veterinaria y Agronomía, Instituto de Ciencias Naturales, Universidad de las Américas, Santiago, Chile
| | - Enrique González-Madrid
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Ma. Andreina Rangel-Ramírez
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Oscar Álvarez-Mardonez
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Omar P. Vallejos
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Constanza Méndez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe Melo-González
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Yorley Duarte
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Ma. Cecilia Opazo
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Medicina Veterinaria y Agronomía, Instituto de Ciencias Naturales, Universidad de las Américas, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A. Riedel
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| |
Collapse
|
34
|
Bester A, O'Brien M, Cotter PD, Dam S, Civai C. Shotgun Metagenomic Sequencing Revealed the Prebiotic Potential of a Fruit Juice Drink with Fermentable Fibres in Healthy Humans. Foods 2023; 12:2480. [PMID: 37444219 DOI: 10.3390/foods12132480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/13/2023] [Accepted: 06/08/2023] [Indexed: 07/15/2023] Open
Abstract
Fibre-based dietary interventions are at the forefront of gut microbiome modulation research, with a wealth of 16S rRNA information to demonstrate the prebiotic effects of isolated fibres. However, there is a distinct lack of data relating to the effect of a combination of soluble and insoluble fibres in a convenient-to-consume fruit juice food matrix on gut microbiota structure, diversity, and function. Here, we aimed to determine the impact of the MOJU Prebiotic Shot, an apple, lemon, ginger, and raspberry fruit juice drink blend containing chicory inulin, baobab, golden kiwi, and green banana powders, on gut microbiota structure and function. Healthy adults (n = 20) were included in a randomised, double-blind, placebo-controlled, cross-over study, receiving 60 mL MOJU Prebiotic Shot or placebo (without the fibre mix) for 3 weeks with a 3-week washout period between interventions. Shotgun metagenomics revealed significant between-group differences in alpha and beta diversity. In addition, the relative abundance of the phyla Actinobacteria and Desulfobacteria was significantly increased as a result of the prebiotic intervention. Nine species were observed to be differentially abundant (uncorrected p-value of <0.05) as a result of the prebiotic treatment. Of these, Bifidobacterium adolescentis and CAG-81 sp900066785 (Lachnospiraceae) were present at increased abundance relative to baseline. Additionally, KEGG analysis showed an increased abundance in pathways associated with arginine biosynthesis and phenylacetate degradation during the prebiotic treatment. Our results show the effects of the daily consumption of 60 mL MOJU Prebiotic Shot for 3 weeks and provide insight into the functional potential of B. adolescentis.
Collapse
Affiliation(s)
- Adri Bester
- London Agri Food Innovation Clinic (LAFIC), School of Applied Sciences, London South Bank University, London SE1 0AA, UK
| | | | | | | | - Claudia Civai
- London Agri Food Innovation Clinic (LAFIC), School of Applied Sciences, London South Bank University, London SE1 0AA, UK
| |
Collapse
|
35
|
Canatar M, Tufan HNG, Ünsal SBE, Koc CY, Ozcan A, Kucuk G, Basmak S, Yatmaz E, Germec M, Yavuz I, Turhan I. Inulinase and fructooligosaccharide production from carob using Aspergillus niger A42 (ATCC 204447) under solid-state fermentation conditions. Int J Biol Macromol 2023:125520. [PMID: 37353118 DOI: 10.1016/j.ijbiomac.2023.125520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/06/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
This study aimed to the production of inulinase and fructooligosaccharides (FOSs) from carob under the solid-state fermentation (SSF) conditions by using Plackett-Burman Design (PBD). Based on the results the maximum inulinase and specific inulinase activities were 249.98 U/mL and 318.29 U/mg protein, respectively. When the fructooligosaccharide (FOS) results were evaluated, the maximum values of 1,1,1-Kestopentaose, 1,1-Kestotetraose, and 1-Kestose were 182.01, 506.16, 132.16 ppm while the lowest and highest total FOS values were 179.35 and 516.66 ppm, respectively. On the other hand, it was observed that the maximum inulinase activity was found at the center points of the design. Therefore, validation fermentations were carried out at center point conditions. Subsequently, the yielded bulk enzyme extracts were partially purified using Spin-X UF membranes with 10, 30, and 50 kDa cut-off values. After purification, the maximum inulinase activity was 247.30 U/mg using a 50 kDa cut-off value. Followed by this process, the purified enzyme was used to produce FOSs and the results indicated that the maximum total FOS amount was 28,712.70 ppm. Consequently, this study successfully demonstrates that Aspergillus niger A42 inulinase produced from carob under the SSF conditions can be used in FOSs production.
Collapse
Affiliation(s)
- Muge Canatar
- Manavgat Vocational School, Akdeniz University, Manavgat, Antalya 07600, Turkey
| | | | | | - Cansu Yılmazer Koc
- Department of Food Engineering, Akdeniz University, Antalya 07058, Turkey
| | - Ali Ozcan
- Department of Food Engineering, Akdeniz University, Antalya 07058, Turkey
| | - Gokce Kucuk
- Department of Food Engineering, Akdeniz University, Antalya 07058, Turkey
| | - Selin Basmak
- Department of Food Engineering, Akdeniz University, Antalya 07058, Turkey
| | - Ercan Yatmaz
- Göynük Culinary Arts Vocational School, Akdeniz University, Kemer, Antalya 07994, Turkey
| | - Mustafa Germec
- Department of Food Engineering, Akdeniz University, Antalya 07058, Turkey
| | - Ibrahim Yavuz
- Technical Sciences Vocational School, Department Of Plant And Animal Production, Organic Agriculture Pr, Akdeniz University, Antalya 07058, Turkey
| | - Irfan Turhan
- Department of Food Engineering, Akdeniz University, Antalya 07058, Turkey.
| |
Collapse
|
36
|
Sheykhsaran E, Abbasi A, Ebrahimzadeh Leylabadlo H, Sadeghi J, Mehri S, Naeimi Mazraeh F, Feizi H, Bannazadeh Baghi H. Gut microbiota and obesity: an overview of microbiota to microbial-based therapies. Postgrad Med J 2023; 99:384-402. [PMID: 35140178 DOI: 10.1136/postgradmedj-2021-141311] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/15/2022] [Indexed: 12/14/2022]
Abstract
The increasing prevalence of obesity and overweight is a significant public concern throughout the world. Obesity is a complex disorder involving an excessive amount of body fat. It is not just a cosmetic concern. It is a medical challenge that increases the risk of other diseases and health circumstances, such as diabetes, heart disease, high blood pressure and certain cancers. Environmental and genetic factors are involved in obesity as a significant metabolic disorder along with diabetes. Gut microbiota (GM) has a high potential for energy harvesting from the diet. In the current review, we aim to consider the role of GM, gut dysbiosis and significant therapies to treat obesity. Dietary modifications, probiotics, prebiotics, synbiotics compounds, using faecal microbiota transplant, and other microbial-based therapies are the strategies to intervene in obesity reducing improvement. Each of these factors serves through various mechanisms including a variety of receptors and compounds to control body weight. Trial and animal investigations have indicated that GM can affect both sides of the energy-balancing equation; first, as an influencing factor for energy utilisation from the diet and also as an influencing factor that regulates the host genes and energy storage and expenditure. All the investigated articles declare the clear and inevitable role of GM in obesity. Overall, obesity and obesity-relevant metabolic disorders are characterised by specific modifications in the human microbiota's composition and functions. The emerging therapeutic methods display positive and promising effects; however, further research must be done to update and complete existing knowledge.
Collapse
Affiliation(s)
- Elham Sheykhsaran
- Immunology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Abbasi
- Student Research Committee, Department of Food Sciences and Technology Research Institute, Faculty of Nutrition Sciences and food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Javid Sadeghi
- Immunology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samaneh Mehri
- Department of Biochemistry and structural Biology, University of Alabama, Birmingham, Alabama, USA
| | - Fariba Naeimi Mazraeh
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Feizi
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Bannazadeh Baghi
- Immunology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
37
|
Knez E, Kadac-Czapska K, Grembecka M. Fermented Vegetables and Legumes vs. Lifestyle Diseases: Microbiota and More. Life (Basel) 2023; 13:life13041044. [PMID: 37109573 PMCID: PMC10141223 DOI: 10.3390/life13041044] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Silages may be preventive against lifestyle diseases, including obesity, diabetes mellitus, or metabolic syndrome. Fermented vegetables and legumes are characterized by pleiotropic health effects, such as probiotic or antioxidant potential. That is mainly due to the fermentation process. Despite the low viability of microorganisms in the gastrointestinal tract, their probiotic potential was confirmed. The modification of microbiota diversity caused by these food products has numerous implications. Most of them are connected to changes in the production of metabolites by bacteria, such as butyrate. Moreover, intake of fermented vegetables and legumes influences epigenetic changes, which lead to inhibition of lipogenesis and decreased appetite. Lifestyle diseases' feature is increased inflammation; thus, foods with high antioxidant potential are recommended. Silages are characterized by having a higher bioavailable antioxidants content than fresh samples. That is due to fermentative microorganisms that produce the enzyme β-glucosidase, which releases these compounds from conjugated bonds with antinutrients. However, fermented vegetables and legumes are rich in salt or salt substitutes, such as potassium chloride. However, until today, silages intake has not been connected to the prevalence of hypertension or kidney failure.
Collapse
Affiliation(s)
- Eliza Knez
- Department of Bromatology, Medical University of Gdańsk, Gen. J. Hallera Aw. 107, 80-416 Gdansk, Poland
| | - Kornelia Kadac-Czapska
- Department of Bromatology, Medical University of Gdańsk, Gen. J. Hallera Aw. 107, 80-416 Gdansk, Poland
| | - Małgorzata Grembecka
- Department of Bromatology, Medical University of Gdańsk, Gen. J. Hallera Aw. 107, 80-416 Gdansk, Poland
| |
Collapse
|
38
|
Ban Q, Chi W, Tan Y, Wang S, Li N, Song L, Huang X, Wang D, Peng W, Granato D, Zhao G. Melatonin improved glucose homeostasis is associated with the reprogrammed gut microbiota and reduced fecal levels of short-chain fatty acids in db/db mice. Food Sci Nutr 2023; 11:2012-2026. [PMID: 37051358 PMCID: PMC10084979 DOI: 10.1002/fsn3.3237] [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: 06/14/2022] [Revised: 11/24/2022] [Accepted: 01/12/2023] [Indexed: 02/01/2023] Open
Abstract
Accumulated evidence shows that melatonin possesses the potential to improve lipid metabolism by modifying gut microbiota and glucose metabolism via regulating the melatonin receptor signaling pathway. However, the contribution of melatonin consumption on glucose homeostasis by affecting gut microbiota has not been investigated in diabetes. In the current work, we investigated the effect of melatonin administration on gut microbiota and glucose homeostasis in db/db mice, a type 2 diabetes model with leptin receptor deficiency. Administration of melatonin through drinking water (at 0.25% and 0.50%) for 12 weeks decreased diabetic polydipsia and polyuria, increased insulin sensitivity and impeded glycemia. The accumulated fecal levels of total short-chain fatty acids (SCFAs) and acetic acid are positively correlated with diabetes-related parameters-homeostasis model assessment of insulin resistance (HOMA-IR) index and fasting blood glucose (FBG) level. The reprogramming of gut microbiota structure and abundance and the reduction of fecal levels of SCFAs, including acetic acid, butyric acid, isovaleric acid, caproic acid, and isobutyric acid, by melatonin may be beneficial for enhancing insulin sensitivity and lowering FBG, which were verified by the results of correlation analysis between acetic acid or total SCFAs and HOMA-IR and FBG. In addition, the melatonin downregulated hepatic genes, including fructose-1,6-bisphosphatase 1, forkhead box O1 alpha, thioredoxin-interacting protein, phosphoenolpyruvate carboxy-kinase (PEPCK), PEPCK1 and a glucose-6-phosphatase catalytic subunit, that responsible for gluconeogenesis support the result that melatonin improved glucose metabolism. Overall, results showed that the melatonin supplementation reduced fecal SCFAs level via reprogramming of gut microbiota, and the reduction of fecal SCFAs level is associated with improved glucose homeostasis in db/db mice.
Collapse
Affiliation(s)
- Qiuyan Ban
- Department of Tea Science, College of HorticultureHenan Agricultural UniversityZhengzhouChina
| | - Wenjing Chi
- Department of Tea Science, College of HorticultureHenan Agricultural UniversityZhengzhouChina
| | - Yu Tan
- Department of Cell Biology, College of Life Science and TechnologyJinan UniversityGuangzhouChina
| | - Shiqiong Wang
- Innovation Team of Food Nutrition and Safety Control, College of Food Science & TechnologyHenan Agricultural UniversityZhengzhouChina
| | - Ning Li
- Innovation Team of Food Nutrition and Safety Control, College of Food Science & TechnologyHenan Agricultural UniversityZhengzhouChina
| | - Lianjun Song
- Innovation Team of Food Nutrition and Safety Control, College of Food Science & TechnologyHenan Agricultural UniversityZhengzhouChina
| | - Xianqing Huang
- Innovation Team of Food Nutrition and Safety Control, College of Food Science & TechnologyHenan Agricultural UniversityZhengzhouChina
| | - Dongxu Wang
- School of Grain Science and TechnologyJiangsu University of Science and TechnologyZhenjiangChina
| | - Wanxi Peng
- Henan Province Engineering Research Center for Biomass Value‐added Products, School of ForestryHenan Agricultural UniversityZhengzhouChina
| | - Daniel Granato
- Bioactivity and Applications Lab, Department of Biological Sciences, Faculty of Science and EngineeringUniversity of LimerickLimerickIreland
| | - Guangshan Zhao
- Innovation Team of Food Nutrition and Safety Control, College of Food Science & TechnologyHenan Agricultural UniversityZhengzhouChina
| |
Collapse
|
39
|
Luo S, Hou Y, Xie L, Zhang H, Liu C, Chen T. Effects of microwave on the potential microbiota modulating effects of agro-industrial by-product fibers among different individuals. Lebensm Wiss Technol 2023. [DOI: 10.1016/j.lwt.2023.114621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
|
40
|
Régnier M, Van Hul M, Roumain M, Paquot A, de Wouters d’Oplinter A, Suriano F, Everard A, Delzenne NM, Muccioli GG, Cani PD. Inulin increases the beneficial effects of rhubarb supplementation on high-fat high-sugar diet-induced metabolic disorders in mice: impact on energy expenditure, brown adipose tissue activity, and microbiota. Gut Microbes 2023; 15:2178796. [PMID: 36803220 PMCID: PMC9980659 DOI: 10.1080/19490976.2023.2178796] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Consumption of prebiotics and plant-based compounds have many beneficial health effects through modulation of gut microbiota composition and are considered as promising nutritional strategy for the treatment of metabolic diseases. In the present study, we assessed the separated and combined effects of inulin and rhubarb on diet-induced metabolic disease in mice. We showed that supplementation with both inulin and rhubarb abolished the total body and fat mass gain upon high-fat and high-sucrose diet (HFHS) as well as several obesity-associated metabolic disorders. These effects were associated with increased energy expenditure, lower whitening of the brown adipose tissue, higher mitochondria activity and increased expression of lipolytic markers in white adipose tissue. Despite modifications of intestinal gut microbiota and bile acid compositions by inulin or rhubarb alone, combination of both inulin and rhubarb had minor additional impact on these parameters. However, the combination of inulin and rhubarb increased the expression of several antimicrobial peptides and higher goblet cell numbers, thereby suggesting a reinforcement of the gut barrier. Together, these results suggest that the combination of inulin and rhubarb in mice potentiates beneficial effects of separated rhubarb and inulin on HFHS-related metabolic disease and could be considered as nutritional strategy for the prevention and treatment of obesity and related pathologies.
Collapse
Affiliation(s)
- Marion Régnier
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium,WELBIO asbl, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Wavre, Belgium
| | - Matthias Van Hul
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium,WELBIO asbl, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Wavre, Belgium
| | - Martin Roumain
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group (BPBL), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Adrien Paquot
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group (BPBL), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Alice de Wouters d’Oplinter
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium,WELBIO asbl, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Wavre, Belgium
| | - Francesco Suriano
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium,WELBIO asbl, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Wavre, Belgium,current address: Institute of Biomedicine, Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Amandine Everard
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium,WELBIO asbl, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Wavre, Belgium
| | - Nathalie M. Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Giulio G. Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group (BPBL), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Patrice D. Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium,WELBIO asbl, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Wavre, Belgium,CONTACT Patrice D. Cani LDRI, Metabolism and Nutrition Research Group, UCLouvain, Université Catholique de Louvain, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Av. E. Mounier, 73 box B1.73.11, B-1200, Brussels, Belgium
| |
Collapse
|
41
|
Ramos Meyers G, Samouda H, Bohn T. Short Chain Fatty Acid Metabolism in Relation to Gut Microbiota and Genetic Variability. Nutrients 2022; 14:5361. [PMID: 36558520 PMCID: PMC9788597 DOI: 10.3390/nu14245361] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
It is widely accepted that the gut microbiota plays a significant role in modulating inflammatory and immune responses of their host. In recent years, the host-microbiota interface has gained relevance in understanding the development of many non-communicable chronic conditions, including cardiovascular disease, cancer, autoimmunity and neurodegeneration. Importantly, dietary fibre (DF) and associated compounds digested by the microbiota and their resulting metabolites, especially short-chain fatty acids (SCFA), were significantly associated with health beneficial effects, such as via proposed anti-inflammatory mechanisms. However, SCFA metabolic pathways are not fully understood. Major steps include production of SCFA by microbiota, uptake in the colonic epithelium, first-pass effects at the liver, followed by biodistribution and metabolism at the host's cellular level. As dietary patterns do not affect all individuals equally, the host genetic makeup may play a role in the metabolic fate of these metabolites, in addition to other factors that might influence the microbiota, such as age, birth through caesarean, medication intake, alcohol and tobacco consumption, pathogen exposure and physical activity. In this article, we review the metabolic pathways of DF, from intake to the intracellular metabolism of fibre-derived products, and identify possible sources of inter-individual variability related to genetic variation. Such variability may be indicative of the phenotypic flexibility in response to diet, and may be predictive of long-term adaptations to dietary factors, including maladaptation and tissue damage, which may develop into disease in individuals with specific predispositions, thus allowing for a better prediction of potential health effects following personalized intervention with DF.
Collapse
Affiliation(s)
- Guilherme Ramos Meyers
- Nutrition and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, 1 A-B, Rue Thomas Edison, 1445 Strassen, Luxembourg
- Doctoral School in Science and Engineering, University of Luxembourg, 2, Avenue de l'Université, 4365 Esch-sur-Alzette, Luxembourg
| | - Hanen Samouda
- Nutrition and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, 1 A-B, Rue Thomas Edison, 1445 Strassen, Luxembourg
| | - Torsten Bohn
- Nutrition and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, 1 A-B, Rue Thomas Edison, 1445 Strassen, Luxembourg
| |
Collapse
|
42
|
Inulin prebiotic dietary supplementation improves metabolic parameters by reducing the Toll-like receptor 4 transmembrane protein gene and interleukin 6 expression in adipose tissue. PHARMANUTRITION 2022. [DOI: 10.1016/j.phanu.2022.100316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
43
|
Ramos-Lopez O. Multi-Omics Nutritional Approaches Targeting Metabolic-Associated Fatty Liver Disease. Genes (Basel) 2022; 13:2142. [PMID: 36421817 PMCID: PMC9690481 DOI: 10.3390/genes13112142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 10/29/2023] Open
Abstract
Currently, metabolic-associated fatty liver disease (MAFLD) is a leading global cause of chronic liver disease, and is expected to become one of the most common indications of liver transplantation. MAFLD is associated with obesity, involving multiple mechanisms such as alterations in lipid metabolism, insulin resistance, hyperinflammation, mitochondrial dysfunction, cell apoptosis, oxidative stress, and extracellular matrix formation. However, the onset and progression of MAFLD is variable among individuals, being influenced by intrinsic (personal) and external environmental factors. In this context, sequence structural variants across the human genome, epigenetic phenomena (i.e., DNA methylation, histone modifications, and long non-coding RNAs) affecting gene expression, gut microbiota dysbiosis, and metabolomics/lipidomic fingerprints may account for differences in MAFLD outcomes through interactions with nutritional features. This knowledge may contribute to gaining a deeper understanding of the molecular and physiological processes underlying MAFLD pathogenesis and phenotype heterogeneity, as well as facilitating the identification of biomarkers of disease progression and therapeutic targets for the implementation of tailored nutritional strategies. This comprehensive literature review highlights the potential of nutrigenetic, nutriepigenetic, nutrimetagenomic, nutritranscriptomics, and nutrimetabolomic approaches for the prevention and management of MAFLD in humans through the lens of precision nutrition.
Collapse
Affiliation(s)
- Omar Ramos-Lopez
- Medicine and Psychology School, Autonomous University of Baja California, Tijuana 22390, Mexico
| |
Collapse
|
44
|
Metabolic Modeling and Bidirectional Culturing of Two Gut Microbes Reveal Cross-Feeding Interactions and Protective Effects on Intestinal Cells. mSystems 2022; 7:e0064622. [PMID: 36005398 PMCID: PMC9600892 DOI: 10.1128/msystems.00646-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The gut microbiota is constituted by thousands of microbial interactions, some of which correspond to the exchange of metabolic by-products or cross-feeding. Inulin and xylan are two major dietary polysaccharides that are fermented by members of the human gut microbiota, resulting in different metabolic profiles. Here, we integrated community modeling and bidirectional culturing assays to study the metabolic interactions between two gut microbes, Phocaeicola dorei and Lachnoclostridium symbiosum, growing in inulin or xylan, and how they provide a protective effect in cultured cells. P. dorei (previously belonging to the Bacteroides genus) was able to consume inulin and xylan, while L. symposium only used certain inulin fractions to produce butyrate as a major end product. Constrained-based flux simulations of refined genome-scale metabolic models of both microbes predicted high lactate and succinate cross-feeding fluxes between P. dorei and L. symbiosum when growing in each fiber. Bidirectional culture assays in both substrates revealed that L. symbiosum growth increased in the presence of P. dorei. Carbohydrate consumption analyses showed a faster carbohydrate consumption in cocultures compared to monocultures. Lactate and succinate concentrations in bidirectional cocultures were lower than in monocultures, pointing to cross-feeding as initially suggested by the model. Butyrate concentrations were similar across all conditions. Finally, supernatants from both bacteria cultured in xylan in bioreactors significantly reduced tumor necrosis factor-α-induced inflammation in HT-29 cells and exerted a protective effect against the TcdB toxin in Caco-2 epithelial cells. Surprisingly, this effect was not observed in inulin cocultures. Overall, these results highlight the predictive value of metabolic models integrated with microbial culture assays for probing microbial interactions in the gut microbiota. They also provide an example of how metabolic exchange could lead to potential beneficial effects in the host. IMPORTANCE Microbial interactions represent the inner connections in the gut microbiome. By integrating mathematical modeling tools and microbial bidirectional culturing, we determined how two gut commensals engage in the exchange of cross-feeding metabolites, lactate and succinate, for increased growth in two fibers. These interactions underpinned butyrate production in cocultures, resulting in a significant reduction in cellular inflammation and protection against microbial toxins when applied to cellular models.
Collapse
|
45
|
Wei S, Wang J, Wang C, Wang Y, Jin M. Inulin mitigates high fructose-induced gut dysbiosis and metabolic dysfunction in mice. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
46
|
Jiang T, Li Y, Li L, Liang T, Du M, Yang L, Yang J, Yang R, Zhao H, Chen M, Ding Y, Zhang J, Wang J, Xie X, Wu Q. Bifidobacterium longum 070103 Fermented Milk Improve Glucose and Lipid Metabolism Disorders by Regulating Gut Microbiota in Mice. Nutrients 2022; 14:nu14194050. [PMID: 36235706 PMCID: PMC9573661 DOI: 10.3390/nu14194050] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 12/08/2022] Open
Abstract
Background: Fermented milk is beneficial for metabolic disorders, while the underlying mechanisms of action remain unclear. This study explored the benefits and underlying mechanisms of Bifidobacterium longum 070103 fermented milk (BLFM) in thirteen-week high-fat and high-sugar (HFHS) fed mice using omics techniques. Methods and results: BLFM with activated glucokinase (GK) was screened by a double-enzyme coupling method. After supplementing BLFM with 10 mL/kg BW per day, fasting blood glucose, total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), and leptin were significantly reduced compared with the HFHS group. Among them, the final body weight (BW), epididymal fat, perirenal fat, and brown fat in BLFM group had better change trends than Lacticaseibacillus rhamnosus GG fermented milk (LGGFM) group. The amplicon and metabolomic data analysis identified Bifibacterium as a key gut microbiota at regulating glycolipid metabolism. BLFM reverses HFHS-induced reduction in bifidobacteria abundance. Further studies showed that BLFM significantly reduces the content of 3-indoxyl sulofphate associated with intestinal barrier damage. In addition, mice treated with BLFM improved BW, glucose tolerance, insulin resistance, and hepatic steatosis. Conclusion: BLFM consumption attenuates obesity and related symptoms in HFHS-fed mice probably via the modulation of gut microbes and metabolites.
Collapse
Affiliation(s)
- Tong Jiang
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Ying Li
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Longyan Li
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Tingting Liang
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Mingzhu Du
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Lingshuang Yang
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Juan Yang
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Runshi Yang
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Hui Zhao
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Moutong Chen
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Yu Ding
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Jumei Zhang
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Juan Wang
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
- Correspondence: (J.W.); (X.X.); (Q.W.)
| | - Xinqiang Xie
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
- Correspondence: (J.W.); (X.X.); (Q.W.)
| | - Qingping Wu
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
- Correspondence: (J.W.); (X.X.); (Q.W.)
| |
Collapse
|
47
|
Almutairi R, Basson AR, Wearsh P, Cominelli F, Rodriguez-Palacios A. Validity of food additive maltodextrin as placebo and effects on human gut physiology: systematic review of placebo-controlled clinical trials. Eur J Nutr 2022; 61:2853-2871. [PMID: 35230477 PMCID: PMC9835112 DOI: 10.1007/s00394-022-02802-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 01/06/2022] [Indexed: 01/16/2023]
Abstract
PURPOSE Maltodextrin (MDX) is a polysaccharide food additive commonly used as oral placebo/control to investigate treatments/interventions in humans. The aims of this study were to appraise the MDX effects on human physiology/gut microbiota, and to assess the validity of MDX as a placebo-control. METHODS We performed a systematic review of randomized-placebo-controlled clinical trials (RCTs) where MDX was used as an orally consumed placebo. Data were extracted from study results where effects (physiological/microbial) were attributed (or not) to MDX, and from study participant outcomes data, before-and-after MDX consumption, for post-publication 're-analysis' using paired-data statistics. RESULTS Of two hundred-sixteen studies on 'MDX/microbiome', seventy RCTs (n = 70) were selected for analysis. Supporting concerns regarding the validity of MDX as a placebo, the majority of RCTs (60%, CI 95% = 0.48-0.76; n = 42/70; Fisher-exact p = 0.001, expected < 5/70) reported MDX-induced physiological (38.1%, n = 16/42; p = 0.005), microbial metabolite (19%, n = 8/42; p = 0.013), or microbiome (50%, n = 21/42; p = 0.0001) effects. MDX-induced alterations on gut microbiome included changes in the Firmicutes and/or Bacteroidetes phyla, and Lactobacillus and/or Bifidobacterium species. Effects on various immunological, inflammatory markers, and gut function/permeability were also documented in 25.6% of the studies (n = 10/42). Notably, there was considerable variability in the direction of effects (decrease/increase), MDX dose, form (powder/pill), duration, and disease/populations studied. Overall, only 20% (n = 14/70; p = 0.026) of studies cross-referenced MDX as a justifiable/innocuous placebo, while 2.9% of studies (n = 2/70) acknowledged their data the opposite. CONCLUSION Orally-consumed MDX often (63.9% of RCTs) induces effects on human physiology/gut microbiota. Such effects question the validity of MDX as a placebo-control in human clinical trials.
Collapse
Affiliation(s)
- Rawan Almutairi
- Department of Pathology, Case Western Reserve University, 2109 Adelbert Road, Cleveland, OH, 44106, USA
| | - Abigail Raffner Basson
- Department of Medicine and Division of Gastroenterology & Liver Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Pamela Wearsh
- Department of Pathology, Case Western Reserve University, 2109 Adelbert Road, Cleveland, OH, 44106, USA
| | - Fabio Cominelli
- Department of Medicine and Division of Gastroenterology & Liver Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- University Hospitals Research and Education Institute, University Hospital Cleveland Medical Center, Cleveland, OH, USA
| | - Alexander Rodriguez-Palacios
- Department of Medicine and Division of Gastroenterology & Liver Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA.
- Germ-Free and Gut Microbiome Core, Cleveland Digestive Diseases Research Core Center, Case Western Reserve University, 2109 Adelbert Road, Cleveland, OH, USA.
- University Hospitals Research and Education Institute, University Hospital Cleveland Medical Center, Cleveland, OH, USA.
| |
Collapse
|
48
|
Olmo R, Wetzels SU, Armanhi JSL, Arruda P, Berg G, Cernava T, Cotter PD, Araujo SC, de Souza RSC, Ferrocino I, Frisvad JC, Georgalaki M, Hansen HH, Kazou M, Kiran GS, Kostic T, Krauss-Etschmann S, Kriaa A, Lange L, Maguin E, Mitter B, Nielsen MO, Olivares M, Quijada NM, Romaní-Pérez M, Sanz Y, Schloter M, Schmitt-Kopplin P, Seaton SC, Selvin J, Sessitsch A, Wang M, Zwirzitz B, Selberherr E, Wagner M. Microbiome Research as an Effective Driver of Success Stories in Agrifood Systems – A Selection of Case Studies. Front Microbiol 2022; 13:834622. [PMID: 35903477 PMCID: PMC9315449 DOI: 10.3389/fmicb.2022.834622] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/24/2022] [Indexed: 12/14/2022] Open
Abstract
Increasing knowledge of the microbiome has led to significant advancements in the agrifood system. Case studies based on microbiome applications have been reported worldwide and, in this review, we have selected 14 success stories that showcase the importance of microbiome research in advancing the agrifood system. The selected case studies describe products, methodologies, applications, tools, and processes that created an economic and societal impact. Additionally, they cover a broad range of fields within the agrifood chain: the management of diseases and putative pathogens; the use of microorganism as soil fertilizers and plant strengtheners; the investigation of the microbial dynamics occurring during food fermentation; the presence of microorganisms and/or genes associated with hazards for animal and human health (e.g., mycotoxins, spoilage agents, or pathogens) in feeds, foods, and their processing environments; applications to improve HACCP systems; and the identification of novel probiotics and prebiotics to improve the animal gut microbiome or to prevent chronic non-communicable diseases in humans (e.g., obesity complications). The microbiomes of soil, plants, and animals are pivotal for ensuring human and environmental health and this review highlights the impact that microbiome applications have with this regard.
Collapse
Affiliation(s)
- Rocío Olmo
- FFoQSI GmbH - Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Tulln, Austria
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
- *Correspondence: Rocío Olmo,
| | - Stefanie Urimare Wetzels
- FFoQSI GmbH - Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Tulln, Austria
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Jaderson Silveira Leite Armanhi
- Symbiomics Microbiome Solutions, Florianópolis, Brazil
- Genomics for Climate Change Research Center, Universidade Estadual de Campinas, Campinas, Brazil
| | - Paulo Arruda
- Genomics for Climate Change Research Center, Universidade Estadual de Campinas, Campinas, Brazil
- Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas, Campinas, Brazil
- Departamento de Genética e Evolução, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, Brazil
| | - Gabriele Berg
- Institute of Environmental Biotechnology, Graz University of Technology, Graz, Austria
- Leibniz Institute for Agricultural Engineering and Bioeconomy (ATB), Potsdam, Germany
- Institute for Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Tomislav Cernava
- Institute of Environmental Biotechnology, Graz University of Technology, Graz, Austria
| | - Paul D. Cotter
- Food Bioscience, Teagasc Food Research Centre Moorepark, Fermoy, Ireland
- APC Microbiome Ireland and VistaMilk, Cork, Ireland
| | - Solon Cordeiro Araujo
- SCA, Consultoria em Microbiologia Agrícola, Campinas, Brazil
- Brazil National Association of Inoculant Producers and Importers (ANPII), Campinas, Brazil
| | - Rafael Soares Correa de Souza
- Symbiomics Microbiome Solutions, Florianópolis, Brazil
- Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas, Campinas, Brazil
| | - Ilario Ferrocino
- Department of Agricultural, Forest and Food Science, University of Torino, Torino, Italy
| | - Jens C. Frisvad
- Department of Biotechnology and Bioengineering, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Marina Georgalaki
- Laboratory of Dairy Research, Department of Food Science and Human Nutrition, Agricultural University of Athens, Athens, Greece
| | - Hanne Helene Hansen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Maria Kazou
- Laboratory of Dairy Research, Department of Food Science and Human Nutrition, Agricultural University of Athens, Athens, Greece
| | | | - Tanja Kostic
- Bioresources Unit, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Tulln, Austria
| | - Susanne Krauss-Etschmann
- Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
- Institute for Experimental Medicine, Christian Albrechts University, Kiel, Germany
| | - Aicha Kriaa
- Microbiota Interaction With Human and Animal Team (MIHA), Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas, France
| | - Lene Lange
- BioEconomy, Research & Advisory, Copenhagen, Denmark
| | - Emmanuelle Maguin
- Microbiota Interaction With Human and Animal Team (MIHA), Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas, France
| | - Birgit Mitter
- Bioresources Unit, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Tulln, Austria
| | - Mette Olaf Nielsen
- Department of Animal Science, Faculty of Technical Sciences, Aarhus University, Tjele, Denmark
| | - Marta Olivares
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Narciso Martín Quijada
- FFoQSI GmbH - Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Tulln, Austria
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Marina Romaní-Pérez
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Yolanda Sanz
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Michael Schloter
- Research Unit Comparative Microbiome Analysis, Helmholtz Center Munich, Neuherberg, Germany
| | | | | | - Joseph Selvin
- School of Life Sciences, Pondicherry University, Puducherry, India
| | - Angela Sessitsch
- Bioresources Unit, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Tulln, Austria
| | - Mengcen Wang
- State Key Laboratory of Rice Biology & Ministry of Agricultural and Rural Affairs Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou, China
| | - Benjamin Zwirzitz
- Institute of Food Science, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Evelyne Selberherr
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Martin Wagner
- FFoQSI GmbH - Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Tulln, Austria
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
49
|
Carneiro PV, Montenegro NDA, Lana A, Amato AA, Santos GM. Lipids from gut microbiota: pursuing a personalized treatment. Trends Mol Med 2022; 28:631-643. [PMID: 35739018 DOI: 10.1016/j.molmed.2022.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 10/18/2022]
Abstract
The discovery of microbiome metabolites has enlivened the field of fecal transplantation for therapeutic purposes. However, the transfer of pathogenic living organisms was recently observed to limit its therapeutic potential by increasing the risk of infection. Lipids produced by gut microbiota enter the circulation and control many phenotypic changes associated with microbiota composition. Fecal lipids significantly impact the regulation of several cell signaling pathways, including inflammation. Focusing on these molecules, we review how bioactive gut microbiota-associated lipids affect cellular functioning and clinical outcome. Here, we interrogate whether the gut microbiota can be considered a cutting-edge biotechnological tool for rapid metabolic engineering of meaningful lipids to offer a novel personalized therapy.
Collapse
Affiliation(s)
- Pamela V Carneiro
- Laboratório de Farmacologia Molecular, Universidade de Brasília, Brasília, Brasil
| | | | - Addison Lana
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, USA
| | - Angelica A Amato
- Laboratório de Farmacologia Molecular, Universidade de Brasília, Brasília, Brasil
| | - Guilherme M Santos
- Laboratório de Farmacologia Molecular, Universidade de Brasília, Brasília, Brasil.
| |
Collapse
|
50
|
Kang JW, Tang X, Walton CJ, Brown MJ, Brewer RA, Maddela RL, Zheng JJ, Agus JK, Zivkovic AM. Multi-Omic Analyses Reveal Bifidogenic Effect and Metabolomic Shifts in Healthy Human Cohort Supplemented With a Prebiotic Dietary Fiber Blend. Front Nutr 2022; 9:908534. [PMID: 35782954 PMCID: PMC9248813 DOI: 10.3389/fnut.2022.908534] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/20/2022] [Indexed: 12/14/2022] Open
Abstract
Dietary fiber, a nutrient derived mainly from whole grains, vegetables, fruits, and legumes, is known to confer a number of health benefits, yet most Americans consume less than half of the daily recommended amount. Convenience and affordability are key factors determining the ability of individuals to incorporate fiber-rich foods into their diet, and many Americans struggle to access, afford, and prepare foods rich in fiber. The objective of this clinical study was to test the changes in microbial community composition, human metabolomics, and general health markers of a convenient, easy to use prebiotic supplement in generally healthy young participants consuming a diet low in fiber. Twenty healthy adults participated in this randomized, placebo-controlled, double-blind, crossover study which was registered at clinicaltrials.gov as NCT03785860. During the study participants consumed 12 g of a prebiotic fiber supplement and 12 g of placebo daily as a powder mixed with water as part of their habitual diet in randomized order for 4 weeks, with a 4-week washout between treatment arms. Fecal microbial DNA was extracted and sequenced by shallow shotgun sequencing on an Illumina NovaSeq. Plasma metabolites were detected using liquid chromatography–mass spectrometry with untargeted analysis. The phylum Actinobacteria, genus Bifidobacterium, and several Bifidobacterium species (B. bifidum, B. adolescentis, B. breve, B. catenulatum, and B. longum) significantly increased after prebiotic supplementation when compared to the placebo. The abundance of genes associated with the utilization of the prebiotic fiber ingredients (sacA, xfp, xpk) and the production of acetate (poxB, ackA) significantly changed with prebiotic supplementation. Additionally, the abundance of genes associated with the prebiotic utilization (xfp, xpk), acetate production (ackA), and choline to betaine oxidation (gbsB) were significantly correlated with changes in the abundance of the genus Bifidobacterium in the prebiotic group. Plasma concentrations of the bacterially produced metabolite indolepropionate significantly increased. The results of this study demonstrate that an easy to consume, low dose (12 g) of a prebiotic powder taken daily increases the abundance of beneficial bifidobacteria and the production of health-promoting bacteria-derived metabolites in healthy individuals with a habitual low-fiber diet.
Collapse
Affiliation(s)
- Jea Woo Kang
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Xinyu Tang
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | | | - Mark J. Brown
- USANA Health Sciences, Inc., Salt Lake City, UT, United States
| | | | | | - Jack Jingyuan Zheng
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Joanne K. Agus
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Angela M. Zivkovic
- Department of Nutrition, University of California, Davis, Davis, CA, United States
- *Correspondence: Angela M. Zivkovic
| |
Collapse
|