1
|
Sun Q, Li BR, Li DH, Wang XY, Wang QY, Jiang ZM, Ning SB, Sun T. WKB ameliorates DSS-induced colitis through inhibiting enteric glial cells activation and altering the intestinal microbiota. J Transl Med 2025; 23:93. [PMID: 39838431 PMCID: PMC11748877 DOI: 10.1186/s12967-025-06085-2] [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: 09/15/2024] [Accepted: 01/06/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a chronic condition influenced by diet, which affects gut microbiota and immune functions. The rising prevalence of IBD, linked to Western diets in developing countries, highlights the need for dietary interventions. This study aimed to assess the impact of white kidney beans (WKB) on gut inflammation and microbiota changes, focusing on their effects on enteric glial cells (EGCs) and immune activity in colitis. METHODS Male C57BL/6 mice were divided into four groups: normal diet (ND), ND with 2.5% dextran sulfate sodium (DSS) for colitis induction, ND with 20% WKB, and WKB with 2.5% DSS. The dietary intervention lasted 17 weeks, with DSS given in the final week. Colonic inflammation was assessed by body weight, disease activity index, and histopathology. Epithelial barrier integrity was evaluated using immunofluorescence, transmission electron microscopy, and permeability assays. EGCs activity was analyzed via immunofluorescence and quantitative real-time PCR. Immune responses were measured using flow cytometry and cytokine profiling, while gut microbiota changes were examined through metagenomic sequencing. RESULTS WKB supplementation significantly alleviated DSS-induced colitis in mice, evidenced by reduced weight loss, disease activity, and improved colonic histology. This effect was linked to enhanced mucosal barrier integrity, seen through increased tight junction protein and Muc2 expression, accompanied by favorable ultrastructural changes. WKB modulated EGCs activity via TNF-like cytokine 1 A inhibition, resulting in reduced glial fibrillary acidic protein expression. Immunologically, it downregulated Th1 and Th17 pro-inflammatory cells, increased Treg cells, and altered cytokine profiles (reduced TNF-α, IFN-γ, IL-17; increased IL-10). Metagenomic analysis showed that WKB restored gut microbiota balance, particularly enhancing beneficial bacteria like Akkermansia. KEGG pathway analysis further indicated that WKB supplementation improved key metabolic pathways, notably those related to phenylalanine, tyrosine, and tryptophan biosynthesis, thereby countering DSS-induced metabolic disruptions. CONCLUSIONS WKB shows promise for treating IBD by enhancing mucosal barriers, inhibiting EGCs activity, balancing Th1/Th17/Treg cells, and restoring gut microbiota and metabolic homeostasis, thereby alleviating colitis symptoms.
Collapse
Affiliation(s)
- Qi Sun
- Department of Gastroenterology, Air Force Medical Center, No. 30 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Bai-Rong Li
- Department of Gastroenterology, Air Force Medical Center, No. 30 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Dong-Hao Li
- Department of Gastroenterology, Air Force Medical Center, No. 30 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Xiao-Ying Wang
- Department of Gastroenterology, Air Force Medical Center, No. 30 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Qian-Yi Wang
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Zhi-Meng Jiang
- Department of Gastroenterology, Air Force Medical Center, No. 30 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Shou-Bin Ning
- Department of Gastroenterology, Air Force Medical Center, No. 30 Fucheng Road, Haidian District, Beijing, 100142, China.
| | - Tao Sun
- Department of Gastroenterology, Air Force Medical Center, No. 30 Fucheng Road, Haidian District, Beijing, 100142, China.
| |
Collapse
|
2
|
Özdemir A, Buyuktuncer Z. Dietary legumes and gut microbiome: a comprehensive review. Crit Rev Food Sci Nutr 2024:1-15. [PMID: 39607793 DOI: 10.1080/10408398.2024.2434725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
The gut microbiome plays a crucial role in human health, affecting metabolic, immune, and cognitive functions. While the impact of various dietary components on the microbiome is well-studied, the effect of legumes remains less explored. This review examines the influence of legume consumption on gut microbiome composition, diversity, and metabolite production, based on 10 human and 21 animal studies. Human studies showed mixed results, with some showing increased microbial diversity and others finding no significant changes. However, legume consumption was linked to increases in beneficial bacteria like Bifidobacterium and Faecalibacterium. Animal studies generally indicated enhanced microbial diversity and composition changes, though these varied by legume type and the host's health. Some studies highlighted legume-induced shifts in bacteria associated with better metabolic health. Overall, the review emphasizes the complexity of legume-microbiome interactions and the need for standardized methodologies and longitudinal studies. While legumes have the potential to positively affect the gut microbiome, the effects are nuanced and depend on context. Future research should investigate the long-term impacts of legume consumption on microbiome stability and its broader health implications, particularly for disease prevention and dietary strategies.
Collapse
Affiliation(s)
- Aslıhan Özdemir
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Hacettepe University, Ankara, Türkiye
| | - Zehra Buyuktuncer
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Hacettepe University, Ankara, Türkiye
| |
Collapse
|
3
|
Mederle AL, Dima M, Stoicescu ER, Căpăstraru BF, Levai CM, Hațegan OA, Maghiari AL. Impact of Gut Microbiome Interventions on Glucose and Lipid Metabolism in Metabolic Diseases: A Systematic Review and Meta-Analysis. Life (Basel) 2024; 14:1485. [PMID: 39598283 PMCID: PMC11595434 DOI: 10.3390/life14111485] [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: 10/14/2024] [Revised: 11/01/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The gut microbiome is increasingly recognized as a key player in metabolic health, influencing glucose and lipid metabolism through various mechanisms. However, the efficacy of gut microbiota-targeted interventions, such as probiotics, prebiotics, fecal microbiota transplantation (FMT), and diet-based treatments, remains unclear for specific metabolic outcomes. In this study, the aim was to evaluate the impact of these interventions on the glucose and lipid parameters in individuals with metabolic diseases such as diabetes mellitus (DM), obesity, and metabolic syndrome. METHODS This systematic review and meta-analysis included 41 randomized controlled trials that investigated the effects of gut microbiota-targeted treatments on metabolic parameters such as fasting glucose, glycated hemoglobin (HbA1c), homeostatic model assessment for insulin resistance (HOMA-IR), total cholesterol, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and triglycerides. A comprehensive search was conducted using databases like PubMed, Google Scholar, and Scopus, focusing on interventions targeting the gut microbiota. A meta-analysis was performed using random-effects models, with effect sizes calculated for each outcome. Risk of bias was assessed using the Cochrane Risk of Bias tool. RESULTS Gut microbiota-targeted interventions significantly reduced fasting glucose, HbA1c, HOMA-IR, total cholesterol, LDL-C, and triglycerides, with moderate heterogeneity observed across studies. The interventions also led to modest increases in HDL-C levels. Probiotic and synbiotic interventions showed the most consistent benefits in improving both glucose and lipid profiles, while FMT yielded mixed results. Short-term interventions showed rapid microbial shifts but less pronounced metabolic improvements, whereas longer-term interventions had more substantial metabolic benefits. CONCLUSIONS In this study, it is demonstrated that gut microbiota-targeted interventions can improve key metabolic outcomes, offering a potential therapeutic strategy for managing metabolic diseases. However, the effectiveness of these interventions varies depending on the type, duration, and population characteristics, highlighting the need for further long-term studies to assess the sustained effects of microbiota modulation on metabolic health.
Collapse
Affiliation(s)
- Alexandra Laura Mederle
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (A.L.M.); (B.F.C.)
| | - Mirabela Dima
- Department of Neonatology, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Emil Robert Stoicescu
- Radiology and Medical Imaging University Clinic, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
- Research Center for Medical Communication, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
- Research Center for Pharmaco-Toxicological Evaluations, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Field of Applied Engineering Sciences, Specialization Statistical Methods and Techniques in Health and Clinical Research, Faculty of Mechanics, “Politehnica” University Timisoara, Mihai Viteazul Boulevard No. 1, 300222 Timisoara, Romania
| | - Bogdan Florin Căpăstraru
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (A.L.M.); (B.F.C.)
- Research Center for Medical Communication, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
| | - Codrina Mihaela Levai
- Research Center for Medical Communication, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
| | - Ovidiu Alin Hațegan
- Discipline of Anatomy and Embriology, Medicine Faculty, “Vasile Goldis” Western University of Arad, Revolution Boulevard 94, 310025 Arad, Romania;
| | - Anca Laura Maghiari
- Department of Anatomy and Embriology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania;
| |
Collapse
|
4
|
Yu H, Tang H, Saxu R, Song Y, Cui X, Xu J, Li N, Cui S, Ge H, Tang W, Gu HF. Effects of Abelmoschus manihot (L.) and its combination with irbesartan in the treatment of diabetic nephropathy via the gut-kidney axis. Front Pharmacol 2024; 15:1424968. [PMID: 39529886 PMCID: PMC11550981 DOI: 10.3389/fphar.2024.1424968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
Background Clinical observations have recently shown that Abelmoschus manihot (L.) in the form of Huangkui capsule (HKC) and in combination with irbesartan (EB) is an effective therapy for diabetic nephropathy (DN) in patients with type 2 diabetes (T2D). The present study aims to explore the mechanisms underlying the therapeutic efficacies of HKC and its combination with EB in DN via the gut-kidney axis. Methods HKC, EB, and their combination or vehicle were administered in db/db mice, which is an animal model for the study of T2D and DN. Comparative analyses of the gut microbiota, serum metabolites, and kidney transcriptomics before and after drug administration were performed. Results After treatment with HKC, EB, and their combination for 4 weeks, the urinary albumin-to-creatinine ratios decreased significantly in the db/db mice with DN. In terms of the gut microbiota, the abundances of Faecalitalea, Blautia, and Streptococcus increased but those of Bacteroidetes, Firmicutes, Enterobacteriaceae, and Desulfovibrio decreased. Parallelly, serum metabolites, mainly including quercetin 3'-glucuronide and L-dopa, were elevated while cortisol and cytochalasin B were reduced. Furthermore, the S100a8, S100a9, Trem1, and Mmp7 genes in the kidneys were downregulated. These altered elements were associated with proteinuria/albuminuria reduction. However, EB had no effects on the changes in blood pressure and specific differentially expressed genes in the kidneys. Conclusion The present study provides experimental evidence that HKC regulates the gut microbiota, circulating metabolites, and renal gene activities, which are useful for better understanding of the action mechanisms of A. manihot in the treatment of DN through the gut-kidney axis.
Collapse
Affiliation(s)
- Hongmei Yu
- Laboratory of Molecular Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Haitao Tang
- Suzhong Pharmaceutical Group Co. Ltd., Suzhong Pharmaceutical Research Institute, Nanjing, Jiangsu Province, China
| | - Rengui Saxu
- Laboratory of Molecular Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Yuhui Song
- Laboratory of Molecular Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Xu Cui
- Laboratory of Molecular Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Jingjing Xu
- Laboratory of Molecular Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Nan Li
- Department of Endocrinology, Jiangsu Province Hospital of Traditional Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Siyuan Cui
- Department of Endocrinology, Wuxi Second People’s Hospital, Jiangnan University Medical Center, Wuxi, Jiangsu Province, China
| | - Haitao Ge
- Suzhong Pharmaceutical Group Co. Ltd., Suzhong Pharmaceutical Research Institute, Nanjing, Jiangsu Province, China
| | - Wei Tang
- Islet Cell Senescence and Function Research Laboratory, Department of Endocrinology, Nanjing Medical University Affiliated Geriatric Hospital/Jiangsu Province Geriatric Hospital, Nanjing, Jiangsu Province, China
| | - Harvest F. Gu
- Laboratory of Molecular Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
5
|
Cronin P, Hurley C, Ryan A, Zamora-Úbeda M, Govindan A, Stanton C, Lane GP, Joyce SA, O’Toole PW, O’Connor EM. Yeast β-glucan supplementation lowers insulin resistance without altering microbiota composition compared with placebo in subjects with type II diabetes: a phase I exploratory study. Br J Nutr 2024; 132:1-12. [PMID: 39439317 PMCID: PMC11617109 DOI: 10.1017/s0007114524002526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 09/21/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
The increased global prevalence of type II diabetes mellitus (T2DM) is associated with consumption of low fibre 'Western diets'. Characteristic metabolic parameters of these individuals include insulin resistance, high fasting and postprandial glucose, as well as low-grade systemic inflammation. Gut microbiota composition is altered significantly in these cohorts suggesting a causative link between diet, microbiota and disease. Dietary fibre consumption has been shown to alleviate these changes and improve glucose parameters in individuals with metabolic disease. We previously reported that yeast β-glucan (yeast beta-1,3/1,6-D-glucan; Wellmune) supplementation ameliorated hyperinsulinaemia and insulin resistance in a murine model. Here, we conducted a randomised, placebo-controlled, two-armed dietary fibre phase I exploratory intervention study in patients with T2DM. The primary outcome measure was alteration to microbiota composition, while the secondary outcome measures included markers of glycaemic control, inflammation as well as metabolomics. Patients were supplemented with 2·5g/day of maltodextrin (placebo) or yeast β-1,3/1,6-D-glucan (treatment). Yeast β-glucan (Wellmune) lowered insulin resistance compared with the placebo maltodextrin after 8 weeks of consumption. TNFα was significantly lower after 4 weeks of β-glucan supplementation. Significantly higher fecal concentrations of several bile acids were detected in the treatment group when compared with the placebo after 8 weeks. These included tauroursodeoxycholic acid, which was previously shown to improve glucose control and lower insulin resistance. Interestingly, the hypoglycaemic and anti-inflammatory effect of yeast β-glucan was independent of any changes in fecal microbiota composition or short-chain fatty acid levels. Our findings highlight the potential of yeast β-glucan to lower insulin resistance in patients with T2DM.
Collapse
Affiliation(s)
- Peter Cronin
- Department of Biological Science, University of Limerick, Limerick, Republic of Ireland
- APC Microbiome Ireland, University College Cork, Cork, Republic of Ireland
| | - Cian Hurley
- School of Microbiology, University College Cork, Cork, Republic of Ireland
| | - Andrew Ryan
- School of Medicine, University of Limerick, Limerick, Republic of Ireland
| | - María Zamora-Úbeda
- APC Microbiome Ireland, University College Cork, Cork, Republic of Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Republic of Ireland
- Teagsac Food Research Centre, Moorepark, Fermoy, Cork, Republic of Ireland
| | - Ashokkumar Govindan
- APC Microbiome Ireland, University College Cork, Cork, Republic of Ireland
- Teagsac Food Research Centre, Moorepark, Fermoy, Cork, Republic of Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Republic of Ireland
- Teagsac Food Research Centre, Moorepark, Fermoy, Cork, Republic of Ireland
| | - Ger P. Lane
- School of Medicine, University of Limerick, Limerick, Republic of Ireland
| | - Susan A. Joyce
- APC Microbiome Ireland, University College Cork, Cork, Republic of Ireland
- School of Biochemistry and Cell Biology, University College Cork, Cork, Republic of Ireland
| | - Paul W. O’Toole
- APC Microbiome Ireland, University College Cork, Cork, Republic of Ireland
- School of Microbiology, University College Cork, Cork, Republic of Ireland
| | - Eibhlís M. O’Connor
- Department of Biological Science, University of Limerick, Limerick, Republic of Ireland
- APC Microbiome Ireland, University College Cork, Cork, Republic of Ireland
- Health Research Institute, University of Limerick, Limerick, Republic of Ireland
| |
Collapse
|
6
|
Zhao J, Fang Z. Alterations of the gut microbiota and metabolites by ShenZhu TiaoPi granule alleviates hyperglycemia in GK rats. Front Microbiol 2024; 15:1420103. [PMID: 39372266 PMCID: PMC11451479 DOI: 10.3389/fmicb.2024.1420103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/29/2024] [Indexed: 10/08/2024] Open
Abstract
ShenZhu TiaoPi granule (STG) is a compound prescription that is used in Chinese medicine for the treatment of type 2 diabetes mellitus (T2DM). Previous studies have indicated a hypoglycaemic effect, but the underlying mechanism remains unclear. Goto-Kakizaki (GK) rats were used to establish an in vivo T2DM model (Mod). The metformin (Met) and STG treatment time was 12 weeks. Fasting blood glucose (FBG) and insulin levels and the area under the glucose curve (GAUC) were measured. Intestinal pathology and permeability were observed. Microbial diversity analysis and metabolomics were used to investigate the underlying mechanisms. Compared with the Con group, the T2DM Mod group presented significant differences in weight, FBG, GAUC, and homeostasis model assessment-insulin resistance (HOMA-IR) indices (p < 0.01). Met and STG improved these indicators (p < 0.01). The pathological morphology and zonula occludens 1 protein levels in the intestines of the Mod group of rats were altered, leading to increases in the lipopolysaccharide (LPS) and interleukin-1β (IL-1β) levels. In the Met and STG groups, the intestinal conditions improved, and the LPS and IL-1β levels significantly decreased (p < 0.01). Changes in the gut microbiota and metabolites occurred in the Mod group. In the STG group, the abundance of Intestinimonas increased, and the abundance of Eubacterium coprostanoligenes decreased significantly (p < 0.05). Moreover, STG also altered 2-deoxyglucose, beta-muricholic acid and dioxolithocholic acid production. In addition, the main metabolic pathways affected by STG were bile acid biosynthesis and cholesterol metabolism. Intestinimonas, D-maltose_and_alpha-lactose may be potential biomarkers for the effects of STG. STG alleviates hyperglycaemia via the gut microbiota and metabolites in GK rats.
Collapse
Affiliation(s)
- Jindong Zhao
- Department of Endocrinology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernizatison of Traditional Chinese Medicine of IHM, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Zhaohui Fang
- Department of Endocrinology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernizatison of Traditional Chinese Medicine of IHM, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
7
|
Jäger R, Abou Sawan S, Purpura M, Grube B, Röske Y, De Costa P, Chong PW. Proprietary alpha-amylase inhibitor formulation from white kidney bean (Phaseolus vulgaris L.) promotes weight and fat loss: a 12-week, double-blind, placebo-controlled, randomized trial. Sci Rep 2024; 14:12685. [PMID: 38830962 PMCID: PMC11148185 DOI: 10.1038/s41598-024-63443-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 05/29/2024] [Indexed: 06/05/2024] Open
Abstract
White kidney bean (Phaseolus vulgaris L.) extracts can aid weight management by reducing calorie intake from complex carbohydrates through alpha-amylase inhibition. We examined the impact of a proprietary aqueous extract from whole dried white kidney beans standardized by its alpha-amylase inhibitor activity (Phase 2 white kidney bean extract (WKBE)) on weight management in subjects with overweight and moderate obesity. In a randomized, double-blind, placebo-controlled fashion, 81 participants completed the study and ingested either a high dose of Phase 2 (1000 mg, WKBE HIGH), a low dose (700 mg, WKBE LOW), or a matching placebo (microcrystalline cellulose, PLA) three times a day, 30 min before meals, for 12 weeks during a calorie restricted diet. In a dose-dependent manner, Phase 2 significantly reduced body weight, fat mass, BMI, waist, hip and in the WKBE HIGH group thigh circumference. Phase 2 is an effective and safe supplement aiding weight and fat loss. ClinicalTrials.gov identifier NCT02930668.
Collapse
|
8
|
Sastre M, Cimbalo A, Mañes J, Manyes L. Gut Microbiota and Nutrition: Strategies for the Prevention and Treatment of Type 2 Diabetes. J Med Food 2024; 27:97-109. [PMID: 38381517 DOI: 10.1089/jmf.2022.0154] [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: 02/22/2024] Open
Abstract
The prevalence of diabetes has increased in last decades worldwide and is expected to continue to do so in the coming years, reaching alarming figures. Evidence have shown that patients with type 2 diabetes (T2D) have intestinal microbial dysbiosis. Moreover, several mechanisms link the microbiota with the appearance of insulin resistance and diabetes. Diet is a crucial factor related to changes in the composition, diversity, and activity of gut microbiota (GM). In this review, the current and future possibilities of nutrient-GM interactions as a strategy to alleviate T2D are discussed, as well as the mechanisms related to decreased low-grade inflammation and insulin resistance. A bibliographic search of clinical trials in Pubmed, Web of Science, and Scopus was carried out, using the terms "gut microbiota, diet and diabetes." The data analyzed in this review support the idea that dietary interventions targeting changes in the microbiota, including the use of prebiotics and probiotics, can improve glycemic parameters. However, these strategies should be individualized taking into account other internal and external factors. Advances in the understanding of the role of the microbiota in the development of metabolic diseases such as T2D, and its translation into a therapeutic approach for the management of diabetes, are necessary to allow a comprehensive approach.
Collapse
Affiliation(s)
- Maria Sastre
- Laboratory of Food Chemistry and Toxicology, University of Valencia, Valencia, Spain
| | - Alessandra Cimbalo
- Laboratory of Food Chemistry and Toxicology, University of Valencia, Valencia, Spain
| | - Jordi Mañes
- Laboratory of Food Chemistry and Toxicology, University of Valencia, Valencia, Spain
| | - Lara Manyes
- Laboratory of Food Chemistry and Toxicology, University of Valencia, Valencia, Spain
| |
Collapse
|
9
|
Feng Q, Niu Z, Zhang S, Wang L, Dong L, Hou D, Zhou S. Protective Effects of White Kidney Bean ( Phaseolus vulgaris L.) against Diet-Induced Hepatic Steatosis in Mice Are Linked to Modification of Gut Microbiota and Its Metabolites. Nutrients 2023; 15:3033. [PMID: 37447359 PMCID: PMC10347063 DOI: 10.3390/nu15133033] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/02/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Disturbances in the gut microbiota and its derived metabolites are closely related to the occurrence and development of hepatic steatosis. The white kidney bean (WKB), as an excellent source of protein, dietary fiber, and phytochemicals, has recently received widespread attention and might exhibit beneficial effects on a high-fat diet (HFD)-induced hepatic steatosis via targeting gut microbiota and its metabolites. The results indicated that HFD, when supplemented with WKB for 12 weeks, could potently reduce obesity symptoms, serum lipid profiles, and glucose, as well as improve the insulin resistance and liver function markers in mice, thereby alleviating hepatic steatosis. An integrated fecal microbiome and metabolomics analysis further demonstrated that WKB was able to normalize HFD-induced gut dysbiosis in mice, thereby mediating the alterations of a wide range of metabolites. Particularly, WKB remarkably increased the relative abundance of probiotics (Akkermansiaceae, Bifidobacteriaceae, and norank_f_Muribaculaceae) and inhibited the growth of hazardous bacteria (Mucispirillum, Enterorhabdus, and Dubosiella) in diet-induced hepatic steatosis mice. Moreover, the significant differential metabolites altered by WKB were annotated in lipid metabolism, which could ameliorate hepatic steatosis via regulating glycerophospholipid metabolism. This study elucidated the role of WKB from the perspective of microbiome and metabolomics in preventing nonalcoholic fatty liver disease, which provides new insights for its application in functional foods.
Collapse
Affiliation(s)
- Qiqian Feng
- School of Food and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China
| | - Zhitao Niu
- School of Food and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China
| | - Siqi Zhang
- School of Food and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China
| | - Li Wang
- School of Food Science and Technology, State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Lijun Dong
- Beijing Yushiyuan Food Co., Ltd., Beijing 101407, China
| | - Dianzhi Hou
- School of Food and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China
| | - Sumei Zhou
- School of Food and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
10
|
Houghton D, Shannon OM, Chater PI, Wilcox MD, Pearson JP, Stanforth K, Jordan C, Avery L, Blain AP, Joel A, Jeffers R, Nolan R, Nelson A, Stewart CJ, Malcomson FC. White kidney bean extract as a nutraceutical: effects on gut microbiota, alpha-amylase inhibition, and user experiences. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2023; 4:e8. [PMID: 39295906 PMCID: PMC11406411 DOI: 10.1017/gmb.2023.5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 05/15/2023] [Accepted: 05/21/2023] [Indexed: 09/21/2024]
Abstract
White kidney bean extract (WKBE) is a nutraceutical often advocated as an anti-obesity agent. The main proposed mechanism for these effects is alpha-amylase inhibition, thereby slowing carbohydrate digestion and absorption. Thus, it is possible that WKBE could impact the gut microbiota and modulate gut health. We investigated the effects of supplementing 20 healthy adults with WKBE for 1 week in a randomised, placebo-controlled crossover trial on the composition of the gut microbiota, gastrointestinal (GI) inflammation (faecal calprotectin), GI symptoms, and stool habits. We conducted in vitro experiments and used a gut model system to explore potential inhibition of alpha-amylase. We gained qualitative insight into participant experiences of using WKBE via focus groups. WKBE supplementation decreased the relative abundance of Bacteroidetes and increased that of Firmicutes, however, there were no significant differences in post-intervention gut microbiota measurements between the WKBE and control. There were no significant effects on GI inflammation or symptoms related to constipation, or stool consistency or frequency. Our in vitro and gut model system analyses showed no effects of WKBE on alpha-amylase activity. Our findings suggest that WKBE may modulate the gut microbiota in healthy adults, however, the underlying mechanism is unlikely due to active site inhibition of alpha-amylase.
Collapse
Affiliation(s)
- David Houghton
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Oliver M Shannon
- Human Nutrition and Exercise Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Peter I Chater
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Matthew D Wilcox
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Jeffrey P Pearson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Kyle Stanforth
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Cara Jordan
- School of Health and Life Sciences, Teesside University, Tees Valley, UK
| | - Leah Avery
- School of Health and Life Sciences, Teesside University, Tees Valley, UK
| | - Alasdair P Blain
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Abraham Joel
- Human Nutrition and Exercise Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Ruth Jeffers
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Ruth Nolan
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Andrew Nelson
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Christopher J Stewart
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Fiona C Malcomson
- Human Nutrition and Exercise Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
11
|
Huang M, Cople-Rodrigues CDS, Waitzberg DL, Rocha IMGD, Curioni CC. Changes in the Gut Microbiota after the Use of Herbal Medicines in Overweight and Obese Individuals: A Systematic Review. Nutrients 2023; 15:2203. [PMID: 37432344 DOI: 10.3390/nu15092203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND Herbal medicine is a low-cost treatment and has been increasingly applied in obesity treatment. Gut microbiota (GM) is strongly associated with obesity pathogenesis. METHODS We conducted a systematic review guided by the question: "Does the use of herbal medicine change the GM composition in obese individuals?" Randomized clinical trials with obese individuals assessing the effects of herbal medicine intervention in GM were retrieved from the Medline, Embase, Scopus, Web of Science, and Cochrane Library databases, including the Cochrane Controlled Trials Register. Two reviewers independently extracted data using standardized piloted data extraction forms and assessed the study-level risk of bias using an Excel template of the Cochrane "Risk of bias" tool 2-RoB 2. RESULTS We identified 1094 articles in the databases. After removing duplicates and reading the title and abstract, 14 publications were fully evaluated, of which seven publications from six studies were considered eligible. The herbs analyzed were Moringa oleifera, Punica granatum, Scutellaria baicalensis, Schisandra chinensis, W-LHIT and WCBE. The analysis showed that Schisandra chinensis and Scutellaria baicalensis had significant effects on weight loss herbal intervention therapy composed by five Chinese herbal medicines Ganoderma lucidum, Coptis chinensis, Astragalus membranaceus, Nelumbo nucifera gaertn, and Fructus aurantii (W-LHIT) and white common bean extract (WCBE) on GM, but no significant changes in anthropometry and laboratory biomarkers. CONCLUSIONS Herbal medicine modulates GM and is associated with increased genera in obese individuals.
Collapse
Affiliation(s)
- Miguel Huang
- Postgraduate Program in Food, Nutrition and Health (PPG-ANS), University of State of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil
| | | | - Dan L Waitzberg
- Department of Gastroenterology, Faculdade de Medicina, LIM-35, Hospital das Clinicas HCFMUSP, School of Medicine, Universidade de São Paulo, São Paulo 05508-090, Brazil
| | - Ilanna Marques Gomes da Rocha
- Department of Gastroenterology, Faculdade de Medicina, LIM-35, Hospital das Clinicas HCFMUSP, School of Medicine, Universidade de São Paulo, São Paulo 05508-090, Brazil
| | - Cintia Chaves Curioni
- Department of Nutrition in Public Health, University of State of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil
| |
Collapse
|