1
|
Zhao X, Qiu Y, Liang L, Fu X. Interkingdom signaling between gastrointestinal hormones and the gut microbiome. Gut Microbes 2025; 17:2456592. [PMID: 39851261 PMCID: PMC11776477 DOI: 10.1080/19490976.2025.2456592] [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: 05/12/2024] [Revised: 08/12/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025] Open
Abstract
The interplay between the gut microbiota and gastrointestinal hormones plays a pivotal role in the health of the host and the development of diseases. As a vital component of the intestinal microecosystem, the gut microbiota influences the synthesis and release of many gastrointestinal hormones through mechanisms such as modulating the intestinal environment, producing metabolites, impacting mucosal barriers, generating immune and inflammatory responses, and releasing neurotransmitters. Conversely, gastrointestinal hormones exert feedback regulation on the gut microbiota by modulating the intestinal environment, nutrient absorption and utilization, and the bacterial biological behavior and composition. The distributions of the gut microbiota and gastrointestinal hormones are anatomically intertwined, and close interactions between the gut microbiota and gastrointestinal hormones are crucial for maintaining gastrointestinal homeostasis. Interventions leveraging the interplay between the gut microbiota and gastrointestinal hormones have been employed in the clinical management of metabolic diseases and inflammatory bowel diseases, such as bariatric surgery and fecal microbiota transplantation, offering promising targets for the treatment of dysbiosis-related diseases.
Collapse
Affiliation(s)
- Xinyu Zhao
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Ye Qiu
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Lanfan Liang
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiangsheng Fu
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Padhi S, Sarkar P, Sahoo D, Rai AK. Potential of fermented foods and their metabolites in improving gut microbiota function and lowering gastrointestinal inflammation. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:4058-4069. [PMID: 38299734 DOI: 10.1002/jsfa.13313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/12/2024] [Accepted: 01/13/2024] [Indexed: 02/02/2024]
Abstract
Foods prepared using microbial conversion of major and minor food components, which are otherwise known as fermented foods continue to impact human health. The live microorganisms and transformed metabolites can also have a deep influence on the gut microbiota, the multifaceted population of microorganisms dwelling inside the gut play a key role in wellbeing of an individual. The probiotic strains delivered through the consumption of fermented food and other bioactive components such as polyphenolic metabolites, bioactive peptides, short-chain fatty acids and others including those produced via gut microbiota mediated transformations have been proposed to balance the gut microbiota diversity and activity, and also to regulate the inflammation in the gut. However, little is known about such effects and only a handful of fermented foods have been explored to date. We herein review the recent knowledge on the dysbiotic gut microbiota linking to major gut inflammatory diseases. Also, evidences that fermented food consumption modulates the gut microbiota, and its impact on the gut inflammation and inflammatory diseases have been discussed. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Srichandan Padhi
- Nutrition Biotechnlogy Division, National Agri-Food Biotechnology Institute, Mohali, India
| | - Puja Sarkar
- Nutrition Biotechnlogy Division, National Agri-Food Biotechnology Institute, Mohali, India
| | | | - Amit Kumar Rai
- Nutrition Biotechnlogy Division, National Agri-Food Biotechnology Institute, Mohali, India
| |
Collapse
|
3
|
Zhang Y, Wang H, Ge Q, Shi J, Zhang H, Gao J, Han J. Polysaccharides of Melientha longistaminea regulates immune function and gut microbiota in cyclophosphamide (CTX)-induced immunosuppressed mice. Int Immunopharmacol 2025; 156:114702. [PMID: 40294471 DOI: 10.1016/j.intimp.2025.114702] [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: 01/10/2025] [Revised: 03/31/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025]
Abstract
MLS is a bioactive extract of Melientha longistaminea shoots. In this study, we characterized the preliminary structure of MLS. And establish in vivo immune model experiments. To investigate the potential mechanism of immunomodulatory effects on CTX-induced immunosuppressed mice. From the experimental results, we found that MLS restored immune organ damage in immunosuppressed mice, increased cytokine and immunoglobulin secretion in the spleen, and attenuated oxidative damage to the liver by CTX. Meanwhile, MLS was also effective in ameliorating immune injury in immunosuppressed mice by activating the NF-κB and Nrf2 signaling pathways. MLS effectively improved the composition of the gut flora and increased the production of SCFAs in the gut. The results indicate that MLS has immunomodulatory effects and provides a research basis for developing Melientha longistaminea polysaccharides as immunomodulatory adjuvants.
Collapse
Affiliation(s)
- Yan Zhang
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China.
| | - Hui Wang
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Qin Ge
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Jing Shi
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Huiying Zhang
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Jiaqi Gao
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Jiaxin Han
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| |
Collapse
|
4
|
Snelson M, Muralitharan RR, Liu CF, Markó L, Forslund SK, Marques FZ, Tang WHW. Gut-Heart Axis: The Role of Gut Microbiota and Metabolites in Heart Failure. Circ Res 2025; 136:1382-1406. [PMID: 40403109 PMCID: PMC12101525 DOI: 10.1161/circresaha.125.325516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 05/24/2025]
Abstract
Heart failure is a global health issue with significant mortality and morbidity. There is increasing evidence that alterations in the gastrointestinal microbiome, gut epithelial permeability, and gastrointestinal disorders contribute to heart failure progression through various pathways, including systemic inflammation, metabolic dysregulation, and modulation of cardiac function. Moreover, several medications used to treat heart failure directly impact the microbiome. The relationship between the gastrointestinal tract and the heart is bidirectional, termed the gut-heart axis. It is increasingly understood that diet-derived microbial metabolites are key mechanistic drivers of the gut-heart axis. This includes, for example, trimethylamine N-oxide and short-chain fatty acids. This review discusses current insights into the interplay between heart failure, its associated risk factors, and the gut microbiome, focusing on key metabolic pathways, the role of dietary interventions, and the potential for gut-targeted therapies. Understanding these complex interactions could pave the way for novel strategies to mitigate heart failure progression and improve patient outcomes.
Collapse
Affiliation(s)
- Matthew Snelson
- Hypertension Research Laboratory, Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Rikeish R. Muralitharan
- Hypertension Research Laboratory, Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Chia-Feng Liu
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland OH, USA
- Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland OH, USA
| | - Lajos Markó
- Charité – Universitätsmedizin Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Experimental and Clinical Research Center ( ECRC), Berlin, Germany
| | - Sofia K. Forslund
- Charité – Universitätsmedizin Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Experimental and Clinical Research Center ( ECRC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Francine Z. Marques
- Hypertension Research Laboratory, Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - W. H. Wilson Tang
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland OH, USA
- Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland OH, USA
| |
Collapse
|
5
|
Turpin W, Lee SH, Croitoru K. Gut Microbiome Signature in Predisease Phase of Inflammatory Bowel Disease: Prediction to Pathogenesis to Prevention. Gastroenterology 2025; 168:902-913. [PMID: 39914464 DOI: 10.1053/j.gastro.2025.01.004] [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: 07/05/2024] [Revised: 01/02/2025] [Accepted: 01/08/2025] [Indexed: 03/23/2025]
Abstract
Advances in understanding the pathogenesis of inflammatory bowel disease (IBD) point toward a key role of the gut microbiome. We review the data describing the changes in the gut microbiome from IBD case-control studies and compare these findings with emerging data from studies of the preclinical phase of IBD. What is apparent is that assessing changes in the composition and function of the gut microbiome during the preclinical phase helps address confounding factors, such as disease activity and drug therapy, which can directly influence the gut microbiome. Understanding these changes in the predisease phase provides a means of predicting IBD in high-risk populations and offers insights into possible mechanisms involved in disease pathogenesis. Finally, we discuss strategies to use this information to design interventions aimed at modulating the microbiome as a means of preventing or delaying the onset of IBD.
Collapse
Affiliation(s)
- Williams Turpin
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Sun-Ho Lee
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Kenneth Croitoru
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
6
|
Su X, Tian Z, Fang Y, Zhou S, Ma S. Effects of high-dose glucocorticoids on gut microbiota in the treatment of Graves' ophthalmopathy. Microbiol Spectr 2025:e0246724. [PMID: 40261021 DOI: 10.1128/spectrum.02467-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 01/14/2025] [Indexed: 04/24/2025] Open
Abstract
Many studies indicate the gut microbiome is associated with diseases caused by administering high-dose glucocorticoids (GCs), such as hypertension, hyperglycemia, and osteoporosis. However, the association between intestinal flora and the use of high-dose GCs remains elusive. We aimed to characterize gut microbiome in Graves' ophthalmopathy (GO) patients after administering high-dose GCs. In this study, 20 primary GO patients were recruited. The differences in gut microbiota of GO patients before and after administering high-dose GCs were analyzed by 16S rDNA sequencing technology. Untargeted metabolomic analysis was used to examine the differences in gut metabolites between two groups. There were significant differences in α and β diversities of gut microbiota in GO patients before and after administering high-dose GCs. The random forest analysis indicated that three intestinal bacteria (Faecalibacterium, Streptococcus, and Prevotella) could distinguish the two groups with the highest accuracy, which was proven by receiver operator characteristic curve and linear discriminant analysis effect size analysis. The short-chain fatty acid-producing ability in GO patients' gut after high-dose GC administration was significantly decreased. The 5-hydroxytryptamine levels significantly increased in the gut of GO patients after administering high-dose GCs. Our study suggests that high-dose GC administration causes the changes in gut microbiome and metabolites. Moreover, the altered flora and metabolites are related to hypertension, hyperglycemia, and osteoporosis. These findings can help understand the development of side effects caused by high-dose GCs and can be further used to develop potential probiotics to facilitate the prevention for those side effects.IMPORTANCEFor the first time, we revealed that gut microbiome and metabolome in Graves' ophthalmopathy patients after high-dose glucocorticoid (GC) administration significantly changed, and the altered flora and metabolites are related to hypertension, hyperglycemia, and osteoporosis. These findings can help understand the development of side effects caused by high-dose GCs and can be further used to develop potential probiotics to facilitate the prevention for those side effects.
Collapse
Affiliation(s)
- Xinhuan Su
- Department of Endocrinology, Department of Geriatrics, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhenyu Tian
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yalun Fang
- Department of Clinical Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Shengnan Zhou
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shizhan Ma
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
7
|
Paniagua G, Couce-Sánchez M, González-Blanco L, Sabater C, García-Fernández A, Rodríguez-Revuelta J, Sáiz PA, Bobes J, Margolles A, García-Portilla MP. Comparative analysis of gut microbiome-derived short-chain fatty acids in patients with severe mental disorder: Insights from schizophrenia and bipolar disorder. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111345. [PMID: 40147807 DOI: 10.1016/j.pnpbp.2025.111345] [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: 12/04/2024] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Both schizophrenia (SZ) and bipolar disorder (BD) are associated with disruptions in the gut microbiome. Short-chain fatty acids (SCFAs), such as acetate, propionate and butyrate, are key metabolites produced by gut bacteria that influence brain function, immune responses and behaviour. We conducted a cross-sectional observational study with 123 patients (86 with SZ and 37 with BD). We analysed faecal samples for SCFA and examined associations with clinical (psychopathology and cognition), metabolic and lifestyle factors. We performed analysis of covariance to find differences in SCFA levels between diagnostic groups, adjusting for covariates. Faecal SCFA levels were numerically higher in the SZ group than in the BD group. However, after adjusting for covariates, a significant sex-by-diagnosis interaction was observed only for acetate levels. Body mass index emerged as a key predictor of SCFA levels but we observed no significant associations with other metabolic or lifestyle variables, including diet, physical activity and blood inflammatory biomarkers. Additionally, SCFA levels showed no correlation with symptom severity or cognitive performance in either group. This study is the first to compare SCFA profiles between SZ and BD, highlighting potential differences in gut microbiota-derived metabolites between these disorders. These findings suggest greater disruption of the gut-brain axis in SZ, potentially reflecting distinct pathophysiological mechanisms involving metabolic and sex-related factors. Further research, including blood SCFA measurements, could better explain the role of SCFAs and explore microbiota-targeted therapeutic strategies for SZ and BD.
Collapse
Affiliation(s)
- Gonzalo Paniagua
- University of Oviedo, Department of Psychiatry, Oviedo, Spain; Servicio de Salud del Principado de Asturias, Psychiatry, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Instituto de Neurociencias del Principado de Asturias (INEUROPA), Oviedo, Spain
| | - Manuel Couce-Sánchez
- University of Oviedo, Department of Psychiatry, Oviedo, Spain; Servicio de Salud del Principado de Asturias, Psychiatry, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Instituto de Neurociencias del Principado de Asturias (INEUROPA), Oviedo, Spain
| | - Leticia González-Blanco
- University of Oviedo, Department of Psychiatry, Oviedo, Spain; Servicio de Salud del Principado de Asturias, Psychiatry, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Instituto de Neurociencias del Principado de Asturias (INEUROPA), Oviedo, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Oviedo, Spain.
| | - Carlos Sabater
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.
| | - Ainoa García-Fernández
- University of Oviedo, Department of Psychiatry, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Instituto de Neurociencias del Principado de Asturias (INEUROPA), Oviedo, Spain
| | - Julia Rodríguez-Revuelta
- University of Oviedo, Department of Psychiatry, Oviedo, Spain; Servicio de Salud del Principado de Asturias, Psychiatry, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Instituto de Neurociencias del Principado de Asturias (INEUROPA), Oviedo, Spain
| | - Pilar A Sáiz
- University of Oviedo, Department of Psychiatry, Oviedo, Spain; Servicio de Salud del Principado de Asturias, Psychiatry, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Instituto de Neurociencias del Principado de Asturias (INEUROPA), Oviedo, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Oviedo, Spain
| | - Julio Bobes
- University of Oviedo, Department of Psychiatry, Oviedo, Spain
| | - Abelardo Margolles
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Instituto de Productos Lácteos de Asturias (IPLA) del Consejo Superior de Investigaciones Científicas (CSIC), Spain
| | - M Paz García-Portilla
- University of Oviedo, Department of Psychiatry, Oviedo, Spain; Servicio de Salud del Principado de Asturias, Psychiatry, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Instituto de Neurociencias del Principado de Asturias (INEUROPA), Oviedo, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Oviedo, Spain
| |
Collapse
|
8
|
Zhang Z, Zhu L, Zhang H, Yu D, Yin Z, Zhan X. Comparative Study on the Effects of Selenium-Enriched Yeasts with Different Selenomethionine Contents on Gut Microbiota and Metabolites. Int J Mol Sci 2025; 26:3315. [PMID: 40244176 PMCID: PMC11989349 DOI: 10.3390/ijms26073315] [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: 01/09/2025] [Revised: 03/29/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Selenium is an essential trace element for human health, but it mainly exists in an inorganic form that cannot be directly absorbed by the body. Brewer's yeast efficiently converts inorganic selenium into bioavailable organic selenium, making selenium-enriched yeast highly significant for human health research. Selenomethionine (SeM) is an important indicator for evaluating the quality of selenium-enriched yeast. Brewer's yeast was selected as the experimental subject, and the digestion of this yeast (Brewer's yeast) was simulated using an in vitro biomimetic gastrointestinal reactor to evaluate the effects of selenium-enriched yeast with various SeM levels on the gut flora of a healthy population. The experimental design comprised normal yeast (control group, OR), yeast containing moderate SeM levels (selenium-enriched group, SE), yeast containing high SeM levels (high-selenium group, MU), and a commercially available group comprising selenium-enriched yeast tablets (MA). The MU group exhibited a significantly higher concentration of short-chain fatty acids than the OR and MA groups during 48 h of fermentation, with significant differences observed (p < 0.05). Sequencing results revealed that the MU group showed significantly increased relative abundances of Bacteroidetes and Actinobacteria, while exhibiting a decreased ratio of Firmicutes to Bacteroidetes, which may simultaneously affect multiple metabolic pathways in vivo. These findings support the theory that selenium-enriched yeast with a high SeM has a more positive effect on human health compared with traditional yeast and offer new ideas for the development and application of selenium-enriched yeast.
Collapse
Affiliation(s)
- Zijian Zhang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China; (Z.Z.); (L.Z.); (H.Z.); (D.Y.); (Z.Y.)
| | - Li Zhu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China; (Z.Z.); (L.Z.); (H.Z.); (D.Y.); (Z.Y.)
- A & F Biotech. Ltd., Burnaby, BC V5A 3P6, Canada
| | - Hongtao Zhang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China; (Z.Z.); (L.Z.); (H.Z.); (D.Y.); (Z.Y.)
| | - Dan Yu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China; (Z.Z.); (L.Z.); (H.Z.); (D.Y.); (Z.Y.)
| | - Zhongwei Yin
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China; (Z.Z.); (L.Z.); (H.Z.); (D.Y.); (Z.Y.)
| | - Xiaobei Zhan
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China; (Z.Z.); (L.Z.); (H.Z.); (D.Y.); (Z.Y.)
| |
Collapse
|
9
|
González-Correa C, Moleón J, Miñano S, Robles-Vera I, de la Visitación N, Guerra-Hernández E, Toral M, Jiménez R, Duarte J, Romero M. Protective Effect of Dietary Fiber on Blood Pressure and Vascular Dysfunction Through Regulation of Sympathetic Tone and Immune Response in Genetic Hypertension. Phytother Res 2025; 39:1858-1875. [PMID: 40122676 DOI: 10.1002/ptr.8484] [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: 11/15/2024] [Revised: 02/17/2025] [Accepted: 02/22/2025] [Indexed: 03/25/2025]
Abstract
The mechanisms underlying the antihypertensive effect of dietary fibers remain poorly understood. This study investigates whether dietary fiber supplementation can prevent cardiovascular damage and high blood pressure in a genetic model of neurogenic hypertension. Six-week-old male spontaneously hypertensive rats (SHR) and their respective normotensive control, Wistar Kyoto rats (WKY), were divided into four groups: Untreated WKY, untreated SHR, SHR treated with resistant starch (SHR + RS), and SHR treated with inulin-type fructans (SHR + ITF) for 12 weeks. Additionally, a faecal microbiota transplantation (FMT) experiment was conducted, transferring faecal content from treated SHR donors to recipient SHRs. A diet rich in RS fiber reduced vascular oxidative stress, inflammation, and high blood pressure. These protective effects were associated with a reshaped gut microbiota, leading to increased short-chain fatty acid production, reduced endotoxemia, decreased sympathetic activity, and a restored balance between Th17 and Treg lymphocytes in mesenteric lymph nodes and aorta. Elevated plasma levels of acetate and butyrate in the SHR + RS group correlated with increased expression of aortic GPR41, GRP43 and PPARδ. Conversely, ITF treatment failed to prevent hypertension or endothelial dysfunction in SHR. FMT from the SHR + RS group to recipient SHR partially replicated these beneficial effects. This study highlights the antihypertensive benefits of dietary insoluble RS fiber, which are attributed to enhanced short-chain fatty acids production in the gut. This leads to improved gut permeability, reduced sympathetic tone, and diminished vascular T-cell accumulation. Therefore, dietary interventions with RS fiber may offer promising therapeutic strategies for preventing hypertension.
Collapse
Affiliation(s)
- Cristina González-Correa
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| | - Javier Moleón
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| | - Sofía Miñano
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Iñaki Robles-Vera
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Néstor de la Visitación
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | | | - Marta Toral
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| | - Rosario Jiménez
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Miguel Romero
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| |
Collapse
|
10
|
Watson JA, Nutten S, Groot A, Hoffmans R, Damen L, Olivier E, Barnett J, Patin A. Safety Assessment of Butyric Acid-Rich Triglyceride Oil: A Novel Palatable Formulation of Butyrate for the Pediatric Population. J Appl Toxicol 2025; 45:587-605. [PMID: 39609950 DOI: 10.1002/jat.4729] [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: 08/16/2024] [Revised: 11/06/2024] [Accepted: 11/06/2024] [Indexed: 11/30/2024]
Abstract
A novel, palatable butyric acid-rich triglyceride oil has been developed and is available as a food supplement for adults in the United States and Canada. A program of safety studies was conducted with butyric acid-rich triglyceride oil for the pediatric population. The oil was tested in a microbial reverse mutation assay Ames Test OECD471 (Organisation for Economic Co-operation and Development) in which all bacterial strains showed negative responses over the complete dose range in two independently repeated experiments. All values were within the laboratory historical control data ranges. Further, data from the human lymphocyte micronucleus assay (OECD487) in the presence or absence of a metabolic activator (S9-mix), the oil did not induce a biologically relevant increase in the number of binucleated cells with micronuclei; therefore, the oil is considered not to be clastogenic or aneugenic. In a 90-day rat repeat dose toxicity study (OECD408), there were no unscheduled deaths, no treatment-related clinical signs, or effects on body weight and body weight gain, food consumption, ophthalmology, FOB parameters (including motor activity), clinical chemistry including thyroid hormones, and sperm parameters. There were no related organ weight changes, macroscopic or microscopic findings. In an extended one-generation reproductive toxicology study (EOGRTS) OECD443, there were no biologically important changes in body weight or body weight gain observed in the P generation male rats during the dosing period. At the end of the dosing period, the mean body weights in the male rats were 98% and 98% of the control group value in the 3720 and 4650 mg/kg/day dose groups, respectively. No biologically important changes in maternal body weights or body weight gains were observed during the premating, gestation, or lactation periods at dose levels up to and including 4650 mg/kg/day. Clinical signs observed in the P generation males and females were within the historical data ranges and not test substance related. There were no test substance-related changes in any other tested outcomes analyzed in the P generation males and females at doses up to and including 4650 mg/kg/day. In the F1 Generation, preweaning clinical signs observed in the males and females were within the historical data ranges and not test article related. There were no statistically significant or biologically relevant abnormalities in any of the parameters analyzed throughout the preweaning period at maternal dose levels up to and including 4650 mg/kg/day. In the postweaning period, there were also no clinical signs observed in males and females; all were within the historical data ranges and not test article related. There were no statistically significant or biologically relevant abnormalities in any of the parameters analyzed throughout the postweaning period at maternal dose levels up to and including 4650 mg/kg/day including body weights. Taken together, data from these toxicity studies show that butyric acid-rich triglyceride oil is extremely safe with a "no observed adverse effect level" (NOAEL) considered to be 4650 mg/kg/day, the highest dose tested.
Collapse
Affiliation(s)
| | | | | | - Roy Hoffmans
- Charles River Laboratories, 's-Hertogenbosch, Netherlands
| | - Lars Damen
- Charles River Laboratories, 's-Hertogenbosch, Netherlands
| | | | - John Barnett
- Charles River Laboratories, Inc, Horsham, Pennsylvania, USA
| | | |
Collapse
|
11
|
Zhang J, Wang X, Fang J, Li Y, Yu Y, Wang J, Sun B. Contributions of Dietary Patterns and Factors to Regulation of Rheumatoid Disease. Int J Mol Sci 2025; 26:2674. [PMID: 40141316 PMCID: PMC11942231 DOI: 10.3390/ijms26062674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease that commonly causes pain in joints and the progressive destruction of cartilage and bone, which significantly reduces the quality of life and increases the social burden. However, there is still no cure for RA, so it is highly important to explore additional adjuvant treatment methods. Studies have indicated that malnutrition, changes in intestinal microbiota, and changes in immune status caused by dietary imbalance are directly related to the onset of RA, indicating that dietary intervention may offer a simple, economical, and practical avenue to relieve RA. Therefore, in this review, we discuss the pathogenesis of RA and summarize the influence of different dietary patterns on RA. In particular, we pointed out that high-fat, high-sugar, and high-salt diets contribute to RA progression, whereas the Mediterranean diet (MD) is beneficial for preventing RA. Furthermore, the ingredients of food, such as dietary fiber, probiotics, and vitamins, help reduce the level of inflammation and relieve joint pain, which may play critical roles in the treatment of RA. Therefore, dietary intervention provides a potential effective approach for adjuvant therapy of RA.
Collapse
Affiliation(s)
- Jingjie Zhang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (J.Z.); (X.W.); (J.F.); (Y.L.); (B.S.)
- China-Canada Joint Laboratory of Food Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing 100048, China
| | - Xueli Wang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (J.Z.); (X.W.); (J.F.); (Y.L.); (B.S.)
- China-Canada Joint Laboratory of Food Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing 100048, China
| | - Juan Fang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (J.Z.); (X.W.); (J.F.); (Y.L.); (B.S.)
- China-Canada Joint Laboratory of Food Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing 100048, China
| | - Yingying Li
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (J.Z.); (X.W.); (J.F.); (Y.L.); (B.S.)
- China-Canada Joint Laboratory of Food Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing 100048, China
| | - Yonghui Yu
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (J.Z.); (X.W.); (J.F.); (Y.L.); (B.S.)
- China-Canada Joint Laboratory of Food Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing 100048, China
| | - Jing Wang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (J.Z.); (X.W.); (J.F.); (Y.L.); (B.S.)
- China-Canada Joint Laboratory of Food Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing 100048, China
| | - Baoguo Sun
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (J.Z.); (X.W.); (J.F.); (Y.L.); (B.S.)
- China-Canada Joint Laboratory of Food Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
12
|
Di Salvo E, Cicero N. Could lactose-free foods influence the gut microbiome? Nat Prod Res 2025; 39:1755-1756. [PMID: 38497246 DOI: 10.1080/14786419.2024.2325584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 02/24/2024] [Indexed: 03/19/2024]
Affiliation(s)
- Eleonora Di Salvo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Nicola Cicero
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| |
Collapse
|
13
|
Tsui Y, Wu X, Zhang X, Peng Y, Mok CKP, Chan FKL, Ng SC, Tun HM. Short-chain fatty acids in viral infection: the underlying mechanisms, opportunities, and challenges. Trends Microbiol 2025; 33:302-320. [PMID: 39505671 DOI: 10.1016/j.tim.2024.10.001] [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: 04/26/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024]
Abstract
Viral infections can cause cellular pathway derangements, cell death, and immunopathological responses, leading to host inflammation. Short-chain fatty acids (SCFAs), produced by the microbiota, have emerged as a potential therapeutic for viral infections due to their ability to modulate these processes. However, SCFAs have been reported to have both beneficial and detrimental effects, necessitating a comprehensive understanding of the underlying mechanisms. This review highlights the complex mechanisms underlying SCFAs' effects on viral infection outcomes. We also emphasize the importance of considering how SCFAs' activities may differ under diverse contexts, including but not limited to target cells with different metabolic wiring, different viral causes of infection, the target organism/cell's nutrient availability and/or energy balance, and hosts with varying microbiome compositions.
Collapse
Affiliation(s)
- Yee Tsui
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xueqi Wu
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xi Zhang
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ye Peng
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Microbiota I-Center (MagIC), Hong Kong, China
| | - Chris Ka Pun Mok
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; S.H. Ho Research Centre for Infectious Diseases, The Chinese University of Hong Kong, Hong Kong, China
| | - Francis K L Chan
- Microbiota I-Center (MagIC), Hong Kong, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China; Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong, China
| | - Siew C Ng
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Microbiota I-Center (MagIC), Hong Kong, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Hein Min Tun
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Microbiota I-Center (MagIC), Hong Kong, China.
| |
Collapse
|
14
|
Farah A, Paul P, Khan AS, Sarkar A, Laws S, Chaari A. Targeting gut microbiota dysbiosis in inflammatory bowel disease: a systematic review of current evidence. Front Med (Lausanne) 2025; 12:1435030. [PMID: 40041456 PMCID: PMC11876558 DOI: 10.3389/fmed.2025.1435030] [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: 05/19/2024] [Accepted: 01/31/2025] [Indexed: 03/06/2025] Open
Abstract
Introduction The dysbiosis of the gut microbiota has been identified as a central factor in the pathogenesis of inflammatory bowel disease (IBD), a chronic condition characterized by frequent recurrence and various adverse effects of traditional therapies. While treatments targeting the gut microbiota show promise, their efficacy in IBD management still requires extensive evaluation. Our systematic review analyzes recent studies to elucidate the advancements and challenges in treating IBD using microbial-based therapies. Methods Through a comprehensive systematic review spanning key scientific databases-PubMed, Embase, Cochrane, Web of Science, Scopus, and Google Scholar-we scrutinized the impact of probiotics, prebiotics, synbiotics, and fecal microbiota transplantation (FMT) on individuals with IBD. Our detailed analysis covered study and participant demographics, along with seven key outcome measures: disease activity index, inflammatory markers, serum cytokines, microbiome composition, adverse effects, and the rates of remission and relapse. Results From 6,080 initial search hits, we included 71 studies that assessed various interventions compared to placebo or standard medical therapy. Although there was notable variation in clinical results while assessing different outcomes, overall, probiotics, prebiotics, and synbiotics enhanced the success rates in inducing remission among IBD patients. Furthermore, we noted significant reductions in levels of pro-inflammatory markers and cytokines. Additionally, the requirement for steroids, hospitalization, and poor outcomes in endoscopic and histological scores were significantly reduced in individuals undergoing FMT. Conclusion Our investigation highlights the potential of targeting gut microbiota dysbiosis with microbial-based therapies in patients with IBD. We recommend conducting larger, placebo-controlled randomized trials with extended follow-up periods to thoroughly assess these treatments' clinical efficacy and safety before widespread recommendations for clinical application.
Collapse
Affiliation(s)
| | | | | | | | | | - Ali Chaari
- Weill Cornell Medicine–Qatar, Qatar Foundation, Education City, Doha, Qatar
| |
Collapse
|
15
|
Khan MM, Kirabo A. Fiber, Fatty Acids, and Blood Pressure: A Gut-Level Solution. Circ Res 2025; 136:358-360. [PMID: 39946444 PMCID: PMC11839179 DOI: 10.1161/circresaha.125.326065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Affiliation(s)
- Mohd Mabood Khan
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 (M.M.K., A.K.)
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 (M.M.K., A.K.)
- Vanderbilt Center for Immunobiology (A.K.)
- Vanderbilt Institute for Infection, Immunology, and Inflammation (A.K.)
- Vanderbilt Institute for Global Health (A.K.)
| |
Collapse
|
16
|
R Muralitharan R, Zheng T, Dinakis E, Xie L, Barbaro-Wahl A, Jama HA, Nakai M, Paterson M, Leung KC, McArdle Z, Mirabito Colafella K, Johnson C, Qin W, Salimova E, Bitto NJ, Kaparakis-Liaskos M, Kaye DM, O'Donnell JA, Mackay CR, Marques FZ. Gut Microbiota Metabolites Sensed by Host GPR41/43 Protect Against Hypertension. Circ Res 2025; 136:e20-e33. [PMID: 39840468 DOI: 10.1161/circresaha.124.325770] [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: 10/25/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Fermentation of dietary fiber by the gut microbiota leads to the production of metabolites called short-chain fatty acids, which lower blood pressure and exert cardioprotective effects. Short-chain fatty acids activate host signaling responses via the functionally redundant receptors GPR41 (G-protein-coupled receptor 41) and GPR43 (G-protein-coupled receptor 43), which are highly expressed by immune cells. Whether and how these receptors protect against hypertension or mediate the cardioprotective effects of dietary fiber remains unknown. METHODS Cardiovascular phenotype was assessed in untreated and Ang II (angiotensin II) treated hypertensive wild-type and GPR41/43 knockout (KO) double knockout male mice fed diets with different levels of fiber content. Some mice received TLR4 (toll-like receptor 4)-antagonist treatment and bone marrow chimeras. SNPs (single-nucleotide polymorphisms) associated with GPR41 and GPR43 expression were assessed in UK Biobank participants. RESULTS Untreated GPR41/43KO mice had unaltered blood pressure but had greater cardiac and renal collagen deposition with higher macrophage numbers in the kidney compared with wild-type mice. Ang II-treated GPR41/43KO mice showed higher systolic blood pressure, cardiorenal weights and collagen deposition, and increased gut permeability, which allows the translocation of gastrointestinal bacterial components such as lipopolysaccharides into the circulation. The use of an antagonist to the lipopolysaccharide receptor, TLR4, a potent proinflammatory signaling molecule, restored the cardiovascular phenotype in GPR41/43KO mice. The lack of GPR41/43 expression in the immune compartment was sufficient to lead to a worsened hypertensive phenotype. We also demonstrate that GPR41/43 is, at least partially, responsible for the blood pressure-lowering and cardioprotective effects of a high-fiber diet. Finally, using the UK Biobank, we provide translational evidence that variants associated with lower expression of both GPR41 and GPR43 are more prevalent in participants with hypertension. CONCLUSIONS Our findings highlight that lack of short-chain fatty acid-receptor signaling via both GPR41 and GPR43 increases risk of high blood pressure, suggesting treatments that target these receptors could be a novel strategy to prevent or treat hypertension.
Collapse
Affiliation(s)
- Rikeish R Muralitharan
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Tenghao Zheng
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Evany Dinakis
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Liang Xie
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia (L.X., C.R.M.)
- Now with Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore (L.X.)
| | - Anastasia Barbaro-Wahl
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Hamdi A Jama
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Michael Nakai
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Madeleine Paterson
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Kwan Charmaine Leung
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Zoe McArdle
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Australia (Z.M., K.M.C.)
| | - Katrina Mirabito Colafella
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Australia (Z.M., K.M.C.)
| | - Chad Johnson
- Bioimaging Platform, La Trobe University, Melbourne, Australia (C.J.)
| | - Wendy Qin
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Ekaterina Salimova
- Monash Bioimaging Facility, Monash University, Melbourne, Australia (E.S.)
| | - Natalie J Bitto
- Department of Microbiology, Anatomy, Physiology and Pharmacology (N.J.B., M. K-L.), La Trobe University, Melbourne, Australia
- La Trobe Research Centre for Extracellular Vesicles (N.J.B., M. K-L.), La Trobe University, Melbourne, Australia
| | - Maria Kaparakis-Liaskos
- Department of Microbiology, Anatomy, Physiology and Pharmacology (N.J.B., M. K-L.), La Trobe University, Melbourne, Australia
- La Trobe Research Centre for Extracellular Vesicles (N.J.B., M. K-L.), La Trobe University, Melbourne, Australia
- Now with Department of Microbiology & Immunology, University of Melbourne, Australia (M. K-L.)
| | - David M Kaye
- Central Clinical School, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Australia (D.M.K.)
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia (D.M.K., F.Z.M.)
- Department of Cardiology, Alfred Hospital, Melbourne, Australia (D.M.K.)
| | - Joanne A O'Donnell
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Charles R Mackay
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia (L.X., C.R.M.)
- School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China (C.R.M.)
| | - Francine Z Marques
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia (D.M.K., F.Z.M.)
| |
Collapse
|
17
|
Cosier DJ, Lambert K, Neale EP, Probst Y, Charlton K. The effect of oral synbiotics on the gut microbiota and inflammatory biomarkers in healthy adults: a systematic review and meta-analysis. Nutr Rev 2025; 83:e4-e24. [PMID: 38341803 PMCID: PMC12086677 DOI: 10.1093/nutrit/nuae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2024] Open
Abstract
CONTEXT Prior research has explored the effect of synbiotics, the combination of probiotics and prebiotics, on the gut microbiota in clinical populations. However, evidence related to the effect of synbiotics on the gut microbiota in healthy adults has not been reviewed to date. OBJECTIVE A systematic review and meta-analysis was conducted to comprehensively investigate the effect of synbiotics on the gut microbiota and inflammatory markers in populations of healthy adults. DATA SOURCES Scopus, PubMed, Web of Science, ScienceDirect, MEDLINE, CINAHL, and The Cochrane Library were systematically searched to retrieve randomized controlled trials examining the primary outcome of gut microbiota or intestinal permeability changes after synbiotic consumption in healthy adults. Secondary outcomes of interest were short-chain fatty acids, inflammatory biomarkers, and gut microbiota diversity. DATA EXTRACTION Weighted (WMD) or standardized mean difference (SMD) outcome data were pooled in restricted maximum likelihood models using random effects. Twenty-seven articles reporting on 26 studies met the eligibility criteria (n = 1319). DATA ANALYSIS Meta-analyses of 16 studies showed synbiotics resulted in a significant increase in Lactobacillus cell count (SMD, 0.74; 95% confidence interval [CI], 0.15, 1.33; P = 0.01) and propionate concentration (SMD, 0.22; 95% CI, 0.02, 0.43; P = 0.03) compared with controls. A trend for an increase in Bifidobacterium relative abundance (WMD, 0.97; 95% CI, 0.42, 2.52; P = 0.10) and cell count (SMD, 0.82; 95% CI, 0.13, 1.88; P = 0.06) was seen. No significant differences in α-diversity, acetate, butyrate, zonulin, IL-6, CRP, or endotoxins were observed. CONCLUSION This review demonstrates that synbiotics modulate the gut microbiota by increasing Lactobacillus and propionate across various healthy adult populations, and may result in increased Bifidobacterium. Significant variations in synbiotic type, dose, and duration should be considered as limitations when applying findings to clinical practice. SYSTEMATIC REVIEW REGISTRATION PROSPERO no. CRD42021284033.
Collapse
Affiliation(s)
- Denelle J Cosier
- School of Medicine, Indigenous and Health Sciences, University of Wollongong, Wollongong, New South Wales, Australia
| | - Kelly Lambert
- School of Medicine, Indigenous and Health Sciences, University of Wollongong, Wollongong, New South Wales, Australia
| | - Elizabeth P Neale
- School of Medicine, Indigenous and Health Sciences, University of Wollongong, Wollongong, New South Wales, Australia
| | - Yasmine Probst
- School of Medicine, Indigenous and Health Sciences, University of Wollongong, Wollongong, New South Wales, Australia
| | - Karen Charlton
- School of Medicine, Indigenous and Health Sciences, University of Wollongong, Wollongong, New South Wales, Australia
| |
Collapse
|
18
|
Yu X, Wu Z, Luo K, Zhou W, Mai K, Zhang W. Comparative analysis of intestinal microbiota and its function on digestion and immunity of juvenile abalone Haliotis discus hannai fed two different sources of dietary soybean protein. FISH & SHELLFISH IMMUNOLOGY 2025; 157:110060. [PMID: 39617308 DOI: 10.1016/j.fsi.2024.110060] [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: 09/24/2024] [Revised: 11/11/2024] [Accepted: 11/28/2024] [Indexed: 12/08/2024]
Abstract
The present study evaluated the replacement of fish meal (FM) with two different soybean protein sources (soybean protein concentrate (SPC) and soybean meal (SBM)) on the intestine microbiota of abalone Haliotis discus hannai and the implications for the host intestinal function and health. The control diet with FM as the main protein source (CON), and the four experimental diets with 50 % and 100 % SBM replacing FM (SBM50 and SBM100), and 25 % and 100 % SPC replacing FM (SPC25 and SPC100) were fed to abalone for 110 days. The intestinal microbiota analysis results revealed that there were no significant differences in α-diversity indices (Chao1, Ace, Sobs, and Shannon) among all groups. However, analysis of similarity (ANOSIM) demonstrated dramatic shifts in the intestinal microbiota component at the genus level among the groups. Venn diagram analysis identified 470 overlapping operational taxonomic units (OTUs) across the five groups, with the SBM50 and CON groups exhibiting the highest and lowest number of unique OTUs, respectively. Firmicutes and Proteobacteria were the predominant phyla in abalone, with Mycoplasma being the dominant genus (CON: 42.95 %; SBM50: 23.98 %; SBM100: 49.32 %; SPC25: 27.20 %; SPC100: 34.25 %). Notably, the pathogens Vibrio abundance in the SPC25 group was significantly lower than in the CON group. The intestinal microbiota networks in the CON, SBM50, SBM100, SPC25, and SPC100 groups consisted of 1757, 2140, 1992, 2281 and 1747 edges, respectively. Furthermore, correlation heatmap results suggested that digestive enzymes and immune indices in abalone were associated with specific intestinal microbiota. Functional prediction via the KEGG pathway analysis revealed that the replacement levels of dietary FM with SBM and SPC significantly affect various biological functions of the intestinal microbiota. In summary, feeding SBM (50 %) and SPC (25 %) diets to abalone increased the abundance of beneficial bacterium in the intestines, contributing to improved digestion and increased growth rate of abalone.
Collapse
Affiliation(s)
- Xiaojun Yu
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), The Key Laboratory of Mariculture (Ministry of Education), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Zhenhua Wu
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), The Key Laboratory of Mariculture (Ministry of Education), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Kai Luo
- Hubei Key Laboratory of Waterlogging Disaster and Agricultural Use of Wetland, Yangtze University, Jingzhou, China
| | - Wanyou Zhou
- Weihai JinPai Biological Technology Co., Ltd, Weihai, 264500, China
| | - Kangsen Mai
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), The Key Laboratory of Mariculture (Ministry of Education), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Wenbing Zhang
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), The Key Laboratory of Mariculture (Ministry of Education), Fisheries College, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
19
|
Marcari AB, Paiva AD, Simon CR, Dos Santos MESM. Leaky Gut Syndrome: An Interplay Between Nutrients and Dysbiosis. Curr Nutr Rep 2025; 14:25. [PMID: 39890659 DOI: 10.1007/s13668-025-00614-7] [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: 01/16/2025] [Indexed: 02/03/2025]
Abstract
PURPOSE OF REVIEW The gut microbiota (GM) is directly related to health and disease. In this context, disturbances resulting from excessive stress, unbalanced diet, alcohol abuse, and antibiotic use, among other factors, can contribute to microbiota imbalance, with significant impacts on host health. This review provides a comprehensive examination of the literature on the influence of diet on dysbiosis and increased intestinal permeability over the past five years. RECENT FINDINGS Diet can be considered one of the main modulating factors of GM, impacting its composition and functionality. Excessive consumption of simple carbohydrates, saturated fats, and processed foods appears to be directly linked to dysbiosis, which can lead to intestinal hyperpermeability and leaky gut syndrome. On the other hand, diets primarily composed of food groups such as nuts, vegetables, fruits, fish, and poultry in moderate quantities, along with limited consumption of red and processed meats, are associated with a more diverse, healthier, and beneficial GM for the host. It is worth noticing that the use of prebiotics and probiotics, omega-3 supplementation, polyunsaturated fatty acids, and vitamins A, B, C, D, and E can positively modulate the intestinal microbiota by altering its metabolic activity, microbial composition, and improve intestinal barrier function. This review points to a new perspective regarding individualized dietary intervention and the need to integrate it into several aspects of cellular biology, biochemistry, and microbiology to prescribe more effective diets and thus contribute to patients' comprehensive health.
Collapse
Affiliation(s)
- Ana Beatriz Marcari
- Department of Biochemistry, Pharmacology and Physiology, Federal University of Triângulo Mineiro, Praça Manoel Terra, 330 - Abadia, Uberaba, MG, 38025-015, Brazil
| | - Aline Dias Paiva
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Praça Manoel Terra, 330 - Abadia, Uberaba, MG, 38025-015, Brazil
| | - Claudio Roberto Simon
- Department of Structural Biology, Federal University of Triângulo Mineiro, Praça Manoel Terra, 330 - Abadia, Uberaba, MG, 38025-015, Brazil
| | - Maria Emilia Soares Martins Dos Santos
- Department of Biochemistry, Pharmacology and Physiology, Federal University of Triângulo Mineiro, Praça Manoel Terra, 330 - Abadia, Uberaba, MG, 38025-015, Brazil.
| |
Collapse
|
20
|
Gomez-Gomez E, Asensio-Grau A, Heredia A, García-Hernández J, Calvo-Lerma J, Andrés A. Screening of the effect of new food prototypes based on fermented lentil and quinoa flours on the colonic microbiota of older adults through a static in vitro colonic fermentation model. Food Funct 2025; 16:570-582. [PMID: 39703012 DOI: 10.1039/d4fo03190c] [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: 12/21/2024]
Abstract
In view of the increasing demand for plant-based protein alternatives, along with the increased protein requirements of older adults, the formulation of new food concepts based on protein-rich ingredients from legumes or pseudo-cereals seems a promising approach. Previous studies have found that solid-state fermentation can improve the nutritional value and digestibility of plant-based commodities; however, scarce evidence exists regarding the effect on prebiotic potential. This study aimed to compare the effect of fermented and unfermented quinoa and lentil flours on the colonic microbiota, as well as that of new food prototypes (gels and breads) made with the flours. After simulating static colonic fermentation of the substrates by using a pool faecal inoculum obtained from four older adults, the microbiota composition (16S rRNA gene sequencing) and short-chain fatty acids (SCFA, gas chromatography GC-FID) were determined. The results showed statistically significant changes in the microbiota at the genus taxonomic level in the range of -5.8% to +17.6% relative abundance. Common findings in all experiments were increased Bacteroides, Acidaminococcus and Parabacteroides and decreased Asteroleplasma, Oscillospiraceae UCG-002 group and Alistipes, as well as increased SCFA production. Overall, the food matrix (flour, gel or bread) was the variable that most affected the changes in the microbiota composition and diversity, while the impact of fermentation of the lentils or quinoa was not statistically significant for most cases. Concretely, the gel-like prototypes produced the most beneficial changes in microbiota composition, while the bread-like formulations were more favourable for increased SCFA and decreased bSCFA production. In conclusion, new food prototypes based on lentil and quinoa flour could have beneficial prebiotic potential. However, few additional advantages were observed from previous solid-state fermentation for obtaining the fermented lentils and quinoa flour.
Collapse
Affiliation(s)
- Elena Gomez-Gomez
- University Institute of Food Engineering (FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain.
| | - Andrea Asensio-Grau
- University Institute of Food Engineering (FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain.
- Research Group In Innovative Technologies for Sustainable Food (ALIOST), Faculty of Pharmacy and Food Sciences, University of Valencia, Avda Vicent Andrés Estellés s/n, 46100 Burjassot, Spain.
| | - Ana Heredia
- University Institute of Food Engineering (FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain.
| | - Jorge García-Hernández
- Advanced Food Microbiology Centre (CAMA), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain.
| | - Joaquim Calvo-Lerma
- University Institute of Food Engineering (FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain.
- Research Group In Innovative Technologies for Sustainable Food (ALIOST), Faculty of Pharmacy and Food Sciences, University of Valencia, Avda Vicent Andrés Estellés s/n, 46100 Burjassot, Spain.
| | - Ana Andrés
- University Institute of Food Engineering (FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain.
| |
Collapse
|
21
|
Shahin NN, Ahmed-Farid OA, Sakr EAE, Kamel EA, Mohamed MM. Oral Supplements of Combined Lactobacillus plantarum and Asparagus officinalis Modulate Gut Microbiota and Alleviate High-Fat Diet-Induced Cognitive Deficits and Neurodegeneration in Rats. Probiotics Antimicrob Proteins 2025:10.1007/s12602-024-10429-7. [PMID: 39777720 DOI: 10.1007/s12602-024-10429-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2024] [Indexed: 01/11/2025]
Abstract
High-fat diet (HFD) consumption disrupts the gut microbiome, instigating metabolic disturbance, brain pathology, and cognitive decline via the gut-brain axis. Probiotic and prebiotic supplementation have been found to improve gut microbiome health, suggesting they could be effective in managing neurodegenerative disorders. This study explored the potential benefits of the probiotic strain Lactobacillus plantarum 20174 (L. plantarum), prebiotic Asparagus officinalis (A. officinalis) extract, or their synbiotic combination against HFD-induced cognitive dysfunction and neurodegeneration in rats. Male Sprague-Dawley rats were fed either a normal diet or an HFD for 24 weeks. Starting from week 13, rats on either diet were divided into vehicle-, prebiotic-, probiotic-, and synbiotic-treated subgroups. Rats received their assigned intervention for 12 more weeks. Prebiotic, probiotic, or synbiotic treatment reverted HFD-instigated alterations in hippocampal amyloid beta, p-tau, α-synuclein, and BDNF levels, leading to restored cognitive function. The tested therapies also improved the HFD-disrupted lipid profile. Interestingly, probiotic and synbiotic therapies attenuated oxidative stress and inflammation, reinstated neurotransmitter balance, and mitigated the energy deficit in HFD-fed rats. Furthermore, L. plantarum and Asparagus administration modulated gut microbiota composition by raising Lactobacillus species and reducing Coliform and Staphylococci bacteria as well as fungi populations. These findings suggest that the oral consumption of A. officinalis prebiotics and/or L. plantarum probiotics alleviates HFD-induced cognitive deficit and neurodegeneration through modulation of the gut-brain axis with superior restorative effects being achieved by synbiotic treatment.
Collapse
Affiliation(s)
- Nancy N Shahin
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | | | - Ebtehag A E Sakr
- Botany Department, Faculty of Women for Arts, Science and Education, Ain Shams University, Cairo, Egypt
| | - Enas A Kamel
- Biochemistry and Nutrition Department, Faculty of Women for Arts Science and Education, Ain Shams University, Cairo, Egypt
| | - Maha M Mohamed
- Home Economic Department, Faculty of Women for Arts Science and Education, Ain Shams University, Cairo, Egypt
| |
Collapse
|
22
|
Sadohara R, Wiesinger JA, Thompson HJ, Glahn RP, Cichy K. Yellow bean ( Phaseolus vulgaris L.) germplasm with less dietary fiber have shorter cooking times and more bioavailable iron. Curr Res Food Sci 2024; 10:100942. [PMID: 39734611 PMCID: PMC11681885 DOI: 10.1016/j.crfs.2024.100942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 12/31/2024] Open
Abstract
Some yellow-colored market classes of dry bean (Phaseolus vulgaris L.) are valued by consumers as an easy-to-digest, fast cooking alternative to darker colored red and black beans, which in comparison generally have longer cooking times and reduced iron bioavailability. There is evidence that the cooking time of yellow beans is linked to the dietary fiber content and may also contribute to nutrient digestibility and bioavailability. Therefore, 52 fast-, moderate-, and slow-cooking yellow beans with diverse iron bioavailability from five market classes (Amarillo, Canario, Green-yellow, Manteca, and Mayocoba) were selected for total dietary fiber (TDF) analysis. TDF was measured as insoluble (IDF) + soluble (SDF) + oligosaccharides (OLIGO) using method AOAC2011.25. Wide variations in the concentrations of IDF (16.0-23.1%), SDF (1.6-7.7%), OLIGO (1.5-3.4%), and TDF (20.6-31.3%) were detected among the yellow beans with various cooking times. Lower concentrations of IDF in yellow beans were associated with shorter cooking times and higher iron bioavailability. The larger sized Andean yellow beans had more SDF than Middle American. One Mayocoba breeding line from Puerto Rico, PR1146-124, had 42% less OLIGOs than average, and may be useful for breeding low-flatulence beans for consumer acceptability. Fast cooking yellow beans provide the same SDF and OLIGO concentrations as yellow beans with longer cooking times but have the added benefit of shorter cooking times (convenience) and provide more bioavailable iron after cooking.
Collapse
Affiliation(s)
- Rie Sadohara
- Department of Plant, Soil and Microbial Sciences, Michigan State University, East Lansing, MI, 48824, USA
| | - Jason A. Wiesinger
- USDA-ARS Robert W. Holley Center for Agriculture and Health, Ithaca, NY, 14853, USA
| | - Henry J. Thompson
- Cancer Prevention Laboratory, Colorado State University, Fort Collins, CO, 80523, USA
| | - Raymond P. Glahn
- USDA-ARS Robert W. Holley Center for Agriculture and Health, Ithaca, NY, 14853, USA
| | - Karen Cichy
- Department of Plant, Soil and Microbial Sciences, Michigan State University, East Lansing, MI, 48824, USA
- USDA-ARS Sugarbeet and Bean Research Unit, East Lansing, MI, 48824, USA
| |
Collapse
|
23
|
Procházková N, Laursen MF, La Barbera G, Tsekitsidi E, Jørgensen MS, Rasmussen MA, Raes J, Licht TR, Dragsted LO, Roager HM. Gut physiology and environment explain variations in human gut microbiome composition and metabolism. Nat Microbiol 2024; 9:3210-3225. [PMID: 39604623 PMCID: PMC11602727 DOI: 10.1038/s41564-024-01856-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 10/11/2024] [Indexed: 11/29/2024]
Abstract
The human gut microbiome is highly personal. However, the contribution of gut physiology and environment to variations in the gut microbiome remains understudied. Here we performed an observational trial using multi-omics to profile microbiome composition and metabolism in 61 healthy adults for 9 consecutive days. We assessed day-to-day changes in gut environmental factors and measured whole-gut and segmental intestinal transit time and pH using a wireless motility capsule in a subset of 50 individuals. We observed substantial daily fluctuations, with intra-individual variations in gut microbiome and metabolism associated with changes in stool moisture and faecal pH, and inter-individual variations accounted for by whole-gut and segmental transit times and pH. Metabolites derived from microbial carbohydrate fermentation correlated negatively with the gut passage time and pH, while proteolytic metabolites and breath methane showed a positive correlation. Finally, we identified associations between segmental transit time/pH and coffee-, diet-, host- and microbial-derived metabolites. Our work suggests that gut physiology and environment are key to understanding the individuality of the human gut microbial composition and metabolism.
Collapse
Affiliation(s)
- Nicola Procházková
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg, Denmark
| | - Martin F Laursen
- National Food Institute, Technical University of Denmark, Kgs Lyngby, Denmark
| | - Giorgia La Barbera
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg, Denmark
| | - Eirini Tsekitsidi
- Laboratory of Hygiene, Social & Preventive Medicine and Medical Statistics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Malte S Jørgensen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg, Denmark
| | - Morten A Rasmussen
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
- Copenhagen Studies on Asthma in Childhood (COPSAC), Herlev-Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Jeroen Raes
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
- Center for Microbiology, VIB, Leuven, Belgium
| | - Tine R Licht
- National Food Institute, Technical University of Denmark, Kgs Lyngby, Denmark
| | - Lars O Dragsted
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg, Denmark
| | - Henrik M Roager
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg, Denmark.
| |
Collapse
|
24
|
Sun G, Li B, Li Y, McClements DJ. Construction of biopolymer-based hydrogel beads for encapsulation, retention, and colonic delivery of tributyrin: Development of functional beverages (fortified bubble tea). Food Res Int 2024; 197:115165. [PMID: 39593376 DOI: 10.1016/j.foodres.2024.115165] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 11/28/2024]
Abstract
Tributyrin (TB) can be hydrolyzed into short chain fatty acids (butyric acid) in the gastrointestinal tract, which are claimed to exhibit beneficial health effects in the colon. However, digestion of tributyrin in the stomach and small intestine may promote its absorption in the upper gastrointestinal tract, thereby reducing its potential colonic health benefits. In this study, we therefore developed a novel method of encapsulating emulsified tributyrin within biopolymer-based hydrogel beads (≈ 800 μm) that were then encapsulated inside the boba beads (≈ 8-10 mm) found in bubble tea. The hydrogel beads were designed to retain and protect the tributyrin under upper gastrointestinal tract (GIT) conditions, but then release it within the colon. The concentration of tributyrin within the boba beads was 33.3 mg/g, which is above the value reported to exhibit health benefits. The morphology, encapsulation properties, water holding capacity, stability, and swelling properties of the tributyrin-loaded boba beads were characterized. Tapioca-based beads exhibited a larger degree of swelling when incubated in water for 12h (>95 %), whereas agar-based beads did not (< 20 %). In addition, the potential gastrointestinal fate of both free and encapsulated tributyrin oil droplets was assessed using an in vitro digestion model. The free tributyrin oil droplets were almost completely hydrolyzed (103.2 %) by the end of the small intestine phase, whereas the tributyrin oil droplets encapsulated within the agar-based (29.4 %) or tapioca-based (40.3 %) boba beads were much more resistant to digestion. The tapioca-based beads were partially broken down as they passed through the simulated GIT, while the agar-based beads maintained their structural integrity. The tapioca-based beads were gradually broken down as they passed through the simulated GIT, while the agar-based ones maintained their structural integrity. Agar beads were also harder, more resilient, and chewier than the tapioca ones. Both types of boba beads tended to swell and disintegrate when heated to high temperatures (90 °C), with the effect being more pronounced for the agar beads. Overall, our results suggest that the agar-based boba beads had greater potential for the delivery of tributyrin to the colon than the tapioca-based ones. The recent popularity of bubble tea means that it may be a suitable vehicle for delivering bioactive food components, like functional lipids, vitamins, nutraceuticals, or probiotics.
Collapse
Affiliation(s)
- Gege Sun
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Bin Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China
| | - Yan Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China
| | | |
Collapse
|
25
|
Guo X, Liu H, Hou R, Chen G, Xiao H, Liu L, Ciftci ON, Liu L. Design strategies of polysaccharide, protein and lipid-based nano-delivery systems in improving the bioavailability of polyphenols and regulating gut homeostasis. Int J Biol Macromol 2024; 283:137463. [PMID: 39547604 DOI: 10.1016/j.ijbiomac.2024.137463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/21/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
Polyphenols are plant secondary metabolites that have attracted much attention due to their anti-inflammatory, antioxidant, and gut homeostasis promoting effects. However, food matrix interaction, poor solubility, and strong digestion and metabolism of polyphenols cause barriers to their absorption in the gastrointestinal tract, which further reduces bioavailability and limits polyphenols' application in the food industry. Nano-delivery systems composed of biocompatible macromolecules (polysaccharides, proteins and lipids) are an effective way to improve the bioavailability of polyphenols. Therefore, this review introduces the construction of biopolymer-based nano-delivery systems and their application in polyphenols, with emphasis on improving the solubility, stability, sustained release and intestinal targeting of polyphenols. In addition, there are possible positive effects of polyphenol-loaded nano-delivery systems on modulating gut microbiota and gut homeostasis, with particular emphasis on modulating intestinal inflammation, metabolic syndrome, and gut-brain axis. It is worth noting that the safety of bio-based nano-delivery systems still need to be further studied. In summary, the application of the bio-based nano-delivery system to deliver polyphenols provides insights for improving the bioavailability of polyphenols and for the treatment of potential diseases in the future.
Collapse
Affiliation(s)
- Xue Guo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, College of Food Science and Technology, Ningbo University, Ningbo, Zhejiang, PR China
| | - Hongyan Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, College of Food Science and Technology, Ningbo University, Ningbo, Zhejiang, PR China
| | - Ruyan Hou
- Anhui Agricultural University, School Tea & Food Science & Technololgy, State Key Lab Tea Plant Biolology & Utilizatilizaytion, Key Lab Food Nutrion & Safety, Hefei 230036, PR China
| | - Guijie Chen
- Anhui Agricultural University, School Tea & Food Science & Technololgy, State Key Lab Tea Plant Biolology & Utilizatilizaytion, Key Lab Food Nutrion & Safety, Hefei 230036, PR China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst 01003, USA
| | - Lingyi Liu
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, USA
| | - Ozan N Ciftci
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, USA
| | - Lianliang Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, College of Food Science and Technology, Ningbo University, Ningbo, Zhejiang, PR China.
| |
Collapse
|
26
|
Aslam H, Lotfaliany M, So D, Berding K, Berk M, Rocks T, Hockey M, Jacka FN, Marx W, Cryan JF, Staudacher HM. Fiber intake and fiber intervention in depression and anxiety: a systematic review and meta-analysis of observational studies and randomized controlled trials. Nutr Rev 2024; 82:1678-1695. [PMID: 38007616 PMCID: PMC11551482 DOI: 10.1093/nutrit/nuad143] [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: 11/27/2023] Open
Abstract
CONTEXT Dietary fibers hold potential to influence depressive and anxiety outcomes by modulating the microbiota-gut-brain axis, which is increasingly recognized as an underlying factor in mental health maintenance. OBJECTIVE Evidence for the effects of fibers on depressive and anxiety outcomes remains unclear. To this end, a systematic literature review and a meta-analysis were conducted that included observational studies and randomized controlled trials (RCTs). DATA SOURCES The PubMed, Embase, CENTRAL, CINAHL, and PsychINFO databases were searched for eligible studies. DATA EXTRACTION Study screening and risk-of-bias assessment were conducted by 2 independent reviewers. DATA ANALYSIS Meta-analyses via random effects models were performed to examine the (1) association between fiber intake and depressive and anxiety outcomes in observational studies, and (2) effect of fiber intervention on depressive and anxiety outcomes compared with placebo in RCTs. A total of 181 405 participants were included in 23 observational studies. In cross-sectional studies, an inverse association was observed between fiber intake and depressive (Cohen's d effect size [d]: -0.11; 95% confidence interval [CI]: -0.16, -0.05) and anxiety (d = -0.25; 95%CI, -0.38, -0.12) outcomes. In longitudinal studies, there was an inverse association between fiber intake and depressive outcomes (d = -0.07; 95%CI, -0.11, -0.04). In total, 740 participants were included in 10 RCTs, all of whom used fiber supplements. Of note, only 1 RCT included individuals with a clinical diagnosis of depression. No difference was found between fiber supplementation and placebo for depressive (d = -0.47; 95%CI, -1.26, 0.31) or anxiety (d = -0.30; 95%CI, -0.67, 0.07) outcomes. CONCLUSION Although observational data suggest a potential benefit for higher fiber intake for depressive and anxiety outcomes, evidence from current RCTs does not support fiber supplementation for improving depressive or anxiety outcomes. More research, including RCTs in clinical populations and using a broad range of fibers, is needed. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42021274898.
Collapse
Affiliation(s)
- Hajara Aslam
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Food & Mood Centre, School of Medicine and Barwon Health, Deakin University, Geelong, Victoria, Australia
| | - Mojtaba Lotfaliany
- IMPACT, School of Medicine and Barwon Health, Deakin University, Geelong, Victoria, Australia
| | - Daniel So
- Department of Gastroenterology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Kirsten Berding
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Michael Berk
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Food & Mood Centre, School of Medicine and Barwon Health, Deakin University, Geelong, Victoria, Australia
- School of Medicine, Deakin University, Geelong, Victoria, Australia
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, Victoria, Australia
- Centre for Youth Mental Health, Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
- Department of Psychiatry, The University of Melbourne, Victoria, Australia
| | - Tetyana Rocks
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Food & Mood Centre, School of Medicine and Barwon Health, Deakin University, Geelong, Victoria, Australia
| | - Meghan Hockey
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Food & Mood Centre, School of Medicine and Barwon Health, Deakin University, Geelong, Victoria, Australia
| | - Felice N Jacka
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Food & Mood Centre, School of Medicine and Barwon Health, Deakin University, Geelong, Victoria, Australia
- Centre for Adolescent Health, Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
- College of Public Health, Medical & Veterinary Sciences, James Cook University, Townsville, Queensland, Australia
| | - Wolfgang Marx
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Food & Mood Centre, School of Medicine and Barwon Health, Deakin University, Geelong, Victoria, Australia
| | - John F Cryan
- School of Medicine, Deakin University, Geelong, Victoria, Australia
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Heidi M Staudacher
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Food & Mood Centre, School of Medicine and Barwon Health, Deakin University, Geelong, Victoria, Australia
| |
Collapse
|
27
|
Beldie LA, Dica CC, Moța M, Pirvu BF, Burticală MA, Mitrea A, Clenciu D, Efrem IC, Vladu BE, Timofticiuc DCP, Roșu MM, Gheonea TC, Amzolini AM, Moța E, Vladu IM. The Interactions Between Diet and Gut Microbiota in Preventing Gestational Diabetes Mellitus: A Narrative Review. Nutrients 2024; 16:4131. [PMID: 39683525 DOI: 10.3390/nu16234131] [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: 10/30/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Recent studies have revealed that dysbiosis, defined as alterations in gut microbiota, plays an important role in the development and the progression of many non-communicable diseases, including metabolic disorders, such as type 2 diabetes mellitus and gestational diabetes mellitus (GDM). The high frequency of GDM makes this disorder an important public health issue, which needs to be addressed in order to reduce both the maternal and fetal complications that are frequently associated with this disease. The studies regarding the connections between gut dysbiosis and GDM are still in their early days, with new research continuously emerging. This narrative review seeks to outline the mechanisms through which a healthy diet that protects the gut microbiota is able to prevent the occurrence of GDM, thus providing medical nutritional therapeutic perspectives for the management of GDM.
Collapse
Affiliation(s)
- Luiza-Andreea Beldie
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Cristina-Camelia Dica
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Maria Moța
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Bianca-Florentina Pirvu
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Marilena-Alexandra Burticală
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Adina Mitrea
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Diana Clenciu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ion Cristian Efrem
- Department of Medical Semiology, Faculty of Dentistry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Beatrice Elena Vladu
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Diana Cristina Protasiewicz Timofticiuc
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Midwives and Nursing, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Maria Magdalena Roșu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Midwives and Nursing, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Theodora Claudia Gheonea
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Anca Maria Amzolini
- Department of Medical Semiology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Eugen Moța
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ionela Mihaela Vladu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
28
|
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
|
29
|
Wang Y, Yuan Z. Gut microbiota in two chickens' breeds: Characteristics and dynamic changes. Microb Pathog 2024; 197:107101. [PMID: 39491567 DOI: 10.1016/j.micpath.2024.107101] [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: 08/10/2024] [Revised: 10/29/2024] [Accepted: 11/02/2024] [Indexed: 11/05/2024]
Abstract
The gut microbiota has been demonstrated to play an important role in host immunity, metabolism, digestion, and growth. However, studies regarding the gut microbiota in Tibetan chickens remains scarce in comparison with other poultry breeds. Here, we investigated the gut microbial characteristics of Tibetan chickens and Arbor Acres broiler chickens (AA broiler chickens) and compare their gut microbial differences. For this purpose, we collected cecal samples from 10 Tibetan chickens and 10 AA broiler chickens for amplicon sequencing. Results indicated that Tibetan chickens exhibited higher gut microbial diversity and abundance compared with AA broiler chickens. Moreover, PCoA-based scatter plot analysis showed that the gut microbial structure of the both breeds was significantly different. Although the dominant bacterial phyla (Firmicutes, Firmicutes and Bacteroidota) of Tibetan chickens and AA broiler chickens were the same, the abundance of some bacterial phyla and genera changed significantly. Microbial taxonomic analysis indicated that the relative abundance of 876 genera of 20 phylum in Tibetan chickens increased significantly, while the relative abundance of 160 genera of 3 phyla decreased significantly compared with AA broiler chickens. In summary, these results indicated that there are significant differences in the gut microbiota between Tibetan chickens and AA broiler chickens. This is an important exploration of the gut microbial characteristics and distribution of Tibetan chickens. The findings may contribute to promoting the development of the Tibetan chicken's industry and reveal the adaptability of Tibetan chickens to the environment.
Collapse
Affiliation(s)
- Yan Wang
- Institute of Animal Husbandry and Veterinary Medicine, Xizang Academy of Agriculture and Animal Husbandry Sciences, Lhasa, 850009, China; Key Laboratory of Livestock and Poultry Genetics and Breeding on Qinghai-Tibet Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, 850009, China
| | - Zhenjie Yuan
- Institute of Animal Husbandry and Veterinary Medicine, Xizang Academy of Agriculture and Animal Husbandry Sciences, Lhasa, 850009, China.
| |
Collapse
|
30
|
Gulliver EL, Di Simone SK, Chonwerawong M, Forster SC. Unlocking the potential for microbiome-based therapeutics to address the sustainable development goal of good health and wellbeing. Microb Biotechnol 2024; 17:e70041. [PMID: 39487814 PMCID: PMC11531172 DOI: 10.1111/1751-7915.70041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/17/2024] [Indexed: 11/04/2024] Open
Abstract
Recent years have witnessed major advances and an ever-growing list of healthcare applications for microbiome-based therapeutics. However, these advances have disproportionately targeted diseases common in high-income countries (HICs). Within low- to middle-income countries (LMIC), opportunities for microbiome-based therapeutics include sexual health epidemics, maternal health, early life mortality, malnutrition, vaccine response and infectious diseases. In this review we detail the advances that have been achieved in microbiome-based therapeutics for these areas of healthcare and identify where further work is required. Current efforts to characterise microbiomes from LMICs will aid in targeting and optimisation of therapeutics and preventative strategies specifically suited to the unmet needs within these populations. Once achieved, opportunities from disease treatment and improved treatment efficacy through to disease prevention and vector control can be effectively addressed using probiotics and live biotherapeutics. Together these strategies have the potential to increase individual health, overcome logistical challenges and reduce overall medical, individual, societal and economic costs.
Collapse
Affiliation(s)
- Emily L. Gulliver
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Molecular and Translational ScienceMonash UniversityClaytonVictoriaAustralia
| | - Sara K. Di Simone
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Ritchie Centre, HudsonInstitute of Medical ResearchMelbourneVictoriaAustralia
- Department of PaediatricsMonash UniversityMelbourneVictoriaAustralia
| | - Michelle Chonwerawong
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Molecular and Translational ScienceMonash UniversityClaytonVictoriaAustralia
| | - Samuel C. Forster
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Molecular and Translational ScienceMonash UniversityClaytonVictoriaAustralia
| |
Collapse
|
31
|
Liang Y, Li Y, Lee C, Yu Z, Chen C, Liang C. Ulcerative colitis: molecular insights and intervention therapy. MOLECULAR BIOMEDICINE 2024; 5:42. [PMID: 39384730 PMCID: PMC11464740 DOI: 10.1186/s43556-024-00207-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: 07/08/2024] [Accepted: 09/13/2024] [Indexed: 10/11/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by abdominal pain, diarrhea, rectal bleeding, and weight loss. The pathogenesis and treatment of UC remain key areas of research interest. Various factors, including genetic predisposition, immune dysregulation, and alterations in the gut microbiota, are believed to contribute to the pathogenesis of UC. Current treatments for UC include 5-aminosalicylic acids, corticosteroids, immunosuppressants, and biologics. However, study reported that the one-year clinical remission rate is only around 40%. It is necessary to prompt the exploration of new treatment modalities. Biologic therapies, such as anti-TNF-α monoclonal antibody and JAK inhibitor, primarily consist of small molecules targeting specific pathways, effectively inducing and maintaining remission. Given the significant role of the gut microbiota, research into intestinal microecologics, such as probiotics and prebiotics, and fecal microbiota transplantation (FMT) shows promising potential in UC treatment. Additionally, medicinal herbs, such as chili pepper and turmeric, used in complementary therapy have shown promising results in UC management. This article reviews recent findings on the mechanisms of UC, including genetic susceptibility, immune cell dynamics and cytokine regulation, and gut microbiota alterations. It also discusses current applications of biologic therapy, herbal therapy, microecologics, and FMT, along with their prospects and challenges.
Collapse
Affiliation(s)
- Yuqing Liang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yang Li
- Department of Respiratory, Sichuan Integrative Medicine Hospital, Chengdu, 610042, China
| | - Chehao Lee
- Department of Traditional Chinese Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ziwei Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Chongli Chen
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Chao Liang
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
32
|
Otaru N, Bajic D, Van den Abbeele P, Vande Velde S, Van Biervliet S, Steinert RE, Rehman A. Bifidogenic Effect of Human Milk Oligosaccharides on Pediatric IBD Fecal Microbiota. Microorganisms 2024; 12:1977. [PMID: 39458287 PMCID: PMC11509818 DOI: 10.3390/microorganisms12101977] [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: 08/07/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
The prevalence of pediatric inflammatory bowel disease (pIBD) has been increasing over the last two decades. Yet, treatment strategies are still limited, in part due to the multifactorial nature of the disease and the complex interplay between genetic, environmental, dietary, immune, and gut microbial factors in its etiology. With their direct and indirect anti-inflammatory properties, human milk oligosaccharides (HMOs) are a promising treatment and management strategy for IBD. However, to date there are no insights into how HMOs may affect pIBD microbiota. Here, we compared the effects of 2'fucosyllactose (2'FL), difucosyllactose (DFL), 3'sialyllactose (3'SL), and blends thereof with fructooligosaccharide (FOS) on microbiota functionality (short- and branched-chain fatty acids, pH, and gas production) and composition (quantitative shallow shotgun sequencing) using fecal material from eight different pediatric Crohn's disease patients inoculated in the SIFR® technology. In general, all HMO treatments significantly increased total short-chain fatty acid production when compared with FOS, despite equal gas production. We found that 2'FL, either alone or in combination with DFL and 3'SL, exhibited a strong acetogenic and propiogenic effect, and 3'SL an acetogenic effect that surpassed the effects observed with FOS. No differences in overall community diversity between HMO- and FOS-treated pIBD microbiota were observed. There was, however, a stronger bifidogenic effect of 2'FL, 3'SL, 2'FL/DFL, and 2'FL/DFL + 3'SL when compared with FOS. In general, 3'SL and HMO blends enriched a broader species profile, including taxa with potentially anti-inflammatory properties, such as Faecalibacterium prausnitzii and Blautia species. This study suggests HMOs as a promising strategy to beneficially alter the gut microbial profile in pIBD.
Collapse
Affiliation(s)
- Nize Otaru
- Health, Nutrition & Care (HNC), DSM-Firmenich, 4303 Kaiseraugst, Switzerland
| | - Danica Bajic
- Health, Nutrition & Care (HNC), DSM-Firmenich, 4303 Kaiseraugst, Switzerland
| | | | - Saskia Vande Velde
- Pediatric Gastroenterology and Nutrition, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Stephanie Van Biervliet
- Pediatric Gastroenterology and Nutrition, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Robert E. Steinert
- Health, Nutrition & Care (HNC), DSM-Firmenich, 4303 Kaiseraugst, Switzerland
| | - Ateequr Rehman
- Health, Nutrition & Care (HNC), DSM-Firmenich, 4303 Kaiseraugst, Switzerland
| |
Collapse
|
33
|
Wang H, Bai J, Miao P, Wei Y, Chen X, Lan H, Qing Y, Zhao M, Li Y, Tang R, Yang X. The key to intestinal health: a review and perspective on food additives. Front Nutr 2024; 11:1420358. [PMID: 39360286 PMCID: PMC11444971 DOI: 10.3389/fnut.2024.1420358] [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: 04/25/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024] Open
Abstract
In this review, we explore the effects of food additives on intestinal health. Food additives, such as preservatives, antioxidants and colorants, are widely used to improve food quality and extend shelf life. However, their effects on intestinal microecology May pose health risks. Starting from the basic functions of food additives and the importance of intestinal microecology, we analyze in detail how additives affect the diversity of intestinal flora, oxidative stress and immune responses. Additionally, we examine the association between food additives and intestinal disorders, including inflammatory bowel disease and irritable bowel syndrome, and how the timing, dosage, and individual differences affect the body's response to additives. We also assess the safety and regulatory policies of food additives and explore the potential of natural additives. Finally, we propose future research directions, emphasizing the refinement of risk assessment methods and the creation of safer, innovative additives.
Collapse
Affiliation(s)
- Haitao Wang
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Junyi Bai
- Chengdu Anorectal Hospital, Chengdu, Sichuan, China
| | - Pengyu Miao
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Yu Wei
- Chengdu Anorectal Hospital, Chengdu, Sichuan, China
| | | | - Haibo Lan
- Chengdu Anorectal Hospital, Chengdu, Sichuan, China
| | - Yong Qing
- Chengdu Anorectal Hospital, Chengdu, Sichuan, China
| | - Meizhu Zhao
- Chengdu Anorectal Hospital, Chengdu, Sichuan, China
| | - Yanyu Li
- Chengdu Anorectal Hospital, Chengdu, Sichuan, China
| | - Rui Tang
- Chengdu Anorectal Hospital, Chengdu, Sichuan, China
| | | |
Collapse
|
34
|
Lu S, Wang C, Ma J, Wang Y. Metabolic mediators: microbial-derived metabolites as key regulators of anti-tumor immunity, immunotherapy, and chemotherapy. Front Immunol 2024; 15:1456030. [PMID: 39351241 PMCID: PMC11439727 DOI: 10.3389/fimmu.2024.1456030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/27/2024] [Indexed: 10/04/2024] Open
Abstract
The human microbiome has recently emerged as a focal point in cancer research, specifically in anti-tumor immunity, immunotherapy, and chemotherapy. This review explores microbial-derived metabolites, emphasizing their crucial roles in shaping fundamental aspects of cancer treatment. Metabolites such as short-chain fatty acids (SCFAs), Trimethylamine N-Oxide (TMAO), and Tryptophan Metabolites take the spotlight, underscoring their diverse origins and functions and their profound impact on the host immune system. The focus is on SCFAs' remarkable ability to modulate immune responses, reduce inflammation, and enhance anti-tumor immunity within the intricate tumor microenvironment (TME). The review critically evaluates TMAO, intricately tied to dietary choices and gut microbiota composition, assessing its implications for cancer susceptibility, progression, and immunosuppression. Additionally, the involvement of tryptophan and other amino acid metabolites in shaping immune responses is discussed, highlighting their influence on immune checkpoints, immunosuppression, and immunotherapy effectiveness. The examination extends to their dynamic interaction with chemotherapy, emphasizing the potential of microbial-derived metabolites to alter treatment protocols and optimize outcomes for cancer patients. A comprehensive understanding of their role in cancer therapy is attained by exploring their impacts on drug metabolism, therapeutic responses, and resistance development. In conclusion, this review underscores the pivotal contributions of microbial-derived metabolites in regulating anti-tumor immunity, immunotherapy responses, and chemotherapy outcomes. By illuminating the intricate interactions between these metabolites and cancer therapy, the article enhances our understanding of cancer biology, paving the way for the development of more effective treatment options in the ongoing battle against cancer.
Collapse
Affiliation(s)
- Shan Lu
- Department of General Practice, The Second Hospital of Jilin University, Changchun, China
| | - Chunling Wang
- Medical Affairs Department, The Second Hospital of Jilin University, Changchun, China
| | - Jingru Ma
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun, China
| | - Yichao Wang
- Department of Obstetrics and Gynecology, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
35
|
Lei L, Qin H, Chen Y, Sun Y, Yin W, Tong S. Association Between Adherence to EAT-Lancet Diet and Risk of Hypertension: An 18-Year National Cohort Study in China. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2024:1-10. [PMID: 39235386 DOI: 10.1080/27697061.2024.2399826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/06/2024]
Abstract
OBJECTIVE The EAT-Lancet Commission has proposed an EAT-Lancet diet (ELD), also known as a planetary health diet (PHD), which is environmentally sustainable and promotes human health. However, the association between this diet and the risk of hypertension remains unclear. This study aimed to determine whether adherence to ELD was associated with a lower risk of hypertension. METHODS 11,402 adults without hypertension at baseline from the China Health and Nutrition Survey were included. The PHD score was used to evaluate ELD adherence, with higher scores reflecting better compliance. Cox proportional hazards regression analysis was utilized to estimate the hazard ratio (HR) with a 95% confidence interval (CI). Additionally, a subgroup analysis was performed to identify the possible effect modifiers, and a mediation analysis was conducted to explore the mediation effects of anthropometric measurements on the association between ELD and hypertension. RESULTS A total of 3993 participants (35%) developed hypertension during 93,058 person-years of follow-up. In the covariate-adjusted model, hypertension risk was reduced in the highest quartile participants compared to the lowest quartile of the PHD score (adjusted HR: 0.79, 95%CI: 0.71-0.87; P-trend < 0.001), which remained significant after sensitivity analysis. Notably, the association was also observed in isolated systolic hypertension, isolated diastolic hypertension, and systolic-diastolic hypertension. Subgroup analysis revealed that the inverse association between the PHD score and hypertension risk was more pronounced in nonsmokers and high-sodium intake consumers than in smokers and low-sodium consumers (P-interaction < 0.05). Additionally, mediation analysis revealed that 23.3% of the association between the PHD score and hypertension risk was mediated by the waist-to-height ratio. CONCLUSION Our findings suggest that a higher adherence to ELD is associated with a lower risk of hypertension. These results emphasize that ELD may serve as a potential strategy to prevent hypertension.
Collapse
Affiliation(s)
- Lifu Lei
- Department of Clinical Nutrition, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haixia Qin
- Department of Clinical Nutrition, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yushi Chen
- Department of Clinical Nutrition, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Sun
- Department of Clinical Nutrition, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wenwei Yin
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Office of Academic Research, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Shiwen Tong
- Department of Clinical Nutrition, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
36
|
Fortuna R, Wang W, Mayengbam S, Tuplin EWN, Sampsell K, Sharkey KA, Hart DA, Reimer RA. Effect of prebiotic fiber on physical function and gut microbiota in adults, mostly women, with knee osteoarthritis and obesity: a randomized controlled trial. Eur J Nutr 2024; 63:2149-2161. [PMID: 38713231 DOI: 10.1007/s00394-024-03415-w] [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: 11/17/2023] [Accepted: 04/18/2024] [Indexed: 05/08/2024]
Abstract
PURPOSE Obesity is a primary risk factor for knee osteoarthritis (OA). Prebiotics enhance beneficial gut microbes and can reduce body fat and inflammation. Our objective was to examine if a 6-month prebiotic intervention improved physical function in adults with knee osteoarthritis and obesity. We also measured knee pain, body composition, quality of life, gut microbiota, inflammatory markers, and serum metabolomics. METHODS Adults (n = 54, mostly women) with co-morbid obesity (BMI > 30 kg/m2) and unilateral/bilateral knee OA were randomly assigned to prebiotic (oligofructose-enriched inulin; 16 g/day; n = 31) or isocaloric placebo (maltodextrin; n = 21) for 6 months. Performance based-tests, knee pain, quality of life, serum metabolomics and inflammatory markers, and fecal microbiota and short-chain fatty acids were assessed. RESULTS Significant between group differences were detected for the change in timed-up-and-go test, 40 m fast paced walk test, and hand grip strength test from baseline that favored prebiotic over placebo. Prebiotic also reduced trunk fat mass (kg) at 6 months and trunk fat (%) at 3 months compared to placebo. There was a trend (p = 0.059) for reduced knee pain at 6 months with prebiotic versus placebo. In gut microbiota analysis, a total of 37 amplicon sequence variants differed between groups. Bifidobacterium abundance was positively correlated with distance walked in the 6-min walk test and hand grip strength. At 6 months, there was a significant separation of serum metabolites between groups with upregulation of phenylalanine and tyrosine metabolism with prebiotic. CONCLUSION Prebiotics may hold promise for conservative management of knee osteoarthritis in adults with obesity and larger trials are warranted. CLINICAL TRIAL REGISTRATION Clinicaltrials.gov/study/NCT04172688.
Collapse
Affiliation(s)
- Rafael Fortuna
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Weilan Wang
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Shyamchand Mayengbam
- Department of Biochemistry, Memorial University of Newfoundland, 45 Artic Ave, St. John's, NL, A1C 5S7, Canada
| | - Erin W Noye Tuplin
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Kara Sampsell
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Keith A Sharkey
- Department of Physiology and Pharmacology, Cumming School of Medicine, Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - David A Hart
- Department of Surgery, and Faculty of Kinesiology, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Raylene A Reimer
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada.
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
37
|
Deleu S, Jacobs I, Vazquez Castellanos JF, Verstockt S, Trindade de Carvalho B, Subotić A, Verstockt B, Arnauts K, Deprez L, Vissers E, Lenfant M, Vandermeulen G, De Hertogh G, Verbeke K, Matteoli G, Huys GRB, Thevelein JM, Raes J, Vermeire S. Effect of Mutant and Engineered High-Acetate-Producing Saccharomyces cerevisiae var. boulardii Strains in Dextran Sodium Sulphate-Induced Colitis. Nutrients 2024; 16:2668. [PMID: 39203805 PMCID: PMC11357622 DOI: 10.3390/nu16162668] [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: 07/02/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
Acetate-producing Saccharomyces cerevisiae var. boulardii strains could exert improved effects on ulcerative colitis, which here, was preclinically evaluated in an acute dextran sodium sulphate induced model of colitis. Nine-week-old female mice were divided into 12 groups, receiving either drinking water or 2.75% dextran sodium sulphate for 7 days, combined with a daily gavage of various treatments with different levels of acetate accumulation: sham control (phosphate buffered saline, no acetate), non-probiotic control (Baker's yeast, no acetate), probiotic control (Enterol®, transient acetate), and additionally several Saccharomyces cerevisiae var. boulardii strains with respectively no, high, and extra-high acetate accumulation. Disease activity was monitored daily, and feces samples were collected at different timepoints. On day 14, the mice were sacrificed, upon which blood and colonic tissue were collected for analysis. Disease activity in inflamed mice was lower when treated with the high-acetate-producing strain compared to sham and non-probiotic controls. The non-acetate-producing strain showed higher disease activity compared to the acetate-producing strains. Accordingly, higher histologic inflammation was observed in non- or transient-acetate-producing strains compared to the sham control, whereas this increase was not observed for high- and extra-high-acetate-producing strains upon induction of inflammation. These anti-inflammatory findings were confirmed by transcriptomic analysis of differentially expressed genes. Moreover, only the strain with the highest acetate production was superior in maintaining a stable gut microbial alpha-diversity upon inflammation. These findings support new possibilities for acetate-mediated management of inflammation in inflammatory bowel disease by administrating high-acetate-producing Saccharomyces cerevisae var. boulardii strains.
Collapse
Affiliation(s)
- Sara Deleu
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Inge Jacobs
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Jorge F. Vazquez Castellanos
- VIB-KU Leuven Center for Microbiology, 3001 Leuven, Belgium (G.R.B.H.)
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, 3000 Leuven, Belgium
| | - Sare Verstockt
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | | | - Ana Subotić
- NovelYeast bv, Bio-Incubator BIO4, Gaston Geenslaan 3, Leuven-Heverlee, 3001 Leuven, Belgium
| | - Bram Verstockt
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
- Department of Gastroenterology and Hepatology, UZ Leuven, KU Leuven, 3000 Leuven, Belgium
| | - Kaline Arnauts
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Lowie Deprez
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Eva Vissers
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Matthias Lenfant
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
- Department of Gastroenterology and Hepatology, UZ Leuven, KU Leuven, 3000 Leuven, Belgium
| | - Greet Vandermeulen
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Gert De Hertogh
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
- Laboratory of Morphology and Molecular Pathology, UZ Leuven, 3000 Leuven, Belgium
| | - Kristin Verbeke
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Gianluca Matteoli
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Geert R. B. Huys
- VIB-KU Leuven Center for Microbiology, 3001 Leuven, Belgium (G.R.B.H.)
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, 3000 Leuven, Belgium
| | - Johan M. Thevelein
- NovelYeast bv, Bio-Incubator BIO4, Gaston Geenslaan 3, Leuven-Heverlee, 3001 Leuven, Belgium
| | - Jeroen Raes
- VIB-KU Leuven Center for Microbiology, 3001 Leuven, Belgium (G.R.B.H.)
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, 3000 Leuven, Belgium
| | - Séverine Vermeire
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
- Department of Gastroenterology and Hepatology, UZ Leuven, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
38
|
Wang Y, Deng H, Xiao L, Pan Y. Escherichia coli Nissle 1917 Protects against Sepsis-Induced Intestinal Damage by Regulating the SCFA/GPRs Signaling Pathway. Microorganisms 2024; 12:1622. [PMID: 39203464 PMCID: PMC11356217 DOI: 10.3390/microorganisms12081622] [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: 06/26/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
This study explores whether Escherichia coli Nissle 1917 (EcN) can preserve the integrity of the intestinal barrier by modulating the metabolism pathway of short-chain fatty acids (SCFAs) in a C57BL/6J mouse model of lipopolysaccharide (LPS)-induced acute enteritis and a model of a Caco-2 monolayer. The study involved establishing a septic shock model in mice through lipopolysaccharide (LPS) injection. Clinical scores and intestinal permeability were meticulously documented. Immunofluorescence was utilized to localize the tight junction proteins. A quantitative real-time polymerase chain reaction (qRT-PCR) was employed to assess the expression of G protein-coupled receptors (GPRs) signaling. Additionally, the supplement of acetate and butyrate with Caco-2 monolayers to elucidate the potential of EcN in augmenting the intestinal barrier primarily via the modulation of SCFAs and qRT-PCR was performed to detect the expression of tight junction proteins and the activation of the GPRs protein signaling pathway. EcN mitigated the clinical symptoms and reduced intestinal permeability in the colon of LPS-induced mice. It also enhanced the production of SCFAs in the gut and upregulated the expression of SCFA receptor proteins GPR41 and GPR43 in the colon tissue. Our findings reveal that EcN activates the SCFA/GPRs pathway, thereby preserving intestinal barrier function and alleviating inflammation in a mouse sepsis model.
Collapse
Affiliation(s)
| | | | | | - Yisheng Pan
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing 100034, China; (Y.W.); (H.D.); (L.X.)
| |
Collapse
|
39
|
Li H, Liu S, Chen H, Zhou L, Chen B, Wang M, Zhang D, Han TL, Zhang H. Gut dysbiosis contributes to SCFAs reduction-associated adipose tissue macrophage polarization in gestational diabetes mellitus. Life Sci 2024; 350:122744. [PMID: 38810793 DOI: 10.1016/j.lfs.2024.122744] [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: 03/22/2024] [Revised: 05/14/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
AIMS The prevalence of gestational diabetes mellitus (GDM) has spurred investigations into various interconnected factors, among which gut dysbiosis is notably prominent. Although gut dysbiosis is strongly associated with GDM, the specific role of the gut microbiome in the pathogenesis of GDM remains unknown. This study aims to explore the pathogenesis of GDM from gut microbiota. MATERIALS AND METHODS In our study, we constructed two GDM mice models: one induced by a high-fat diet (HFD) and the other through fecal microbiota transplantation (FMT) from GDM patients. In vitro, we used a co-culture system of RAW264.7 and 3T3-L1 adipocytes. KEY FINDINGS We induced a GDM-like state in pregnant mice by FMT from GDM patients, which was consistent with the HFD model. A potential mechanism identified involves the diminished abundance of SCFA-producing microbiota, which reduces SCFAs, particularly propionic acid and butyric acid. In vitro, butyric and propionic acids were observed to alleviate LPS-induced TLR4-NF-κB activation, thereby reducing inflammation levels and inhibiting adipose insulin resistance via the PI3K/AKT signaling pathway. This reduction appears to trigger the polarization of adipose tissue macrophages toward M1 and promote insulin resistance in adipose tissue. SIGNIFICANCE Our study fills this knowledge gap by finding that alterations in gut microbiota have an independent impact on hyperglycemia and insulin resistance in the GDM state. In vivo and in vitro, gut dysbiosis is linked to adipose tissue inflammation and insulin resistance via the bacterial product SCFAs in the GDM state, providing new insights into the pathogenesis of GDM.
Collapse
Affiliation(s)
- Hongli Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; State Key Laboratory of Maternal and Fetal Medicine of Chongqing, Chongqing Medical University, Chongqing 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, Chongqing Medical University, Chongqing 400016, China
| | - Siqi Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; State Key Laboratory of Maternal and Fetal Medicine of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Hao Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; State Key Laboratory of Maternal and Fetal Medicine of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Linwei Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; State Key Laboratory of Maternal and Fetal Medicine of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Bingnan Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Min Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Dan Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ting-Li Han
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Hua Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; State Key Laboratory of Maternal and Fetal Medicine of Chongqing, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
40
|
Chang CCJ, Liu B, Liebmann JM, Cioffi GA, Winn BJ. Glaucoma and the Human Microbiome. J Glaucoma 2024; 33:529-538. [PMID: 38809163 DOI: 10.1097/ijg.0000000000002448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/11/2024] [Indexed: 05/30/2024]
Abstract
PURPOSE OF REVIEW To explore a view of the human microbiome as an interconnected, functional, dynamic system that may be linked to the pathogenesis and progression of glaucoma. METHODS A literature review was undertaken that included publications from 1966 to 2023. RESULTS Bacterial lipopolysaccharides (LPS) activate toll-like receptors (TLR) and mediate the human immune response. The LPS-TLR4 pathway is a potential avenue for the ocular, gut, and oral microbiomes to interface and/or influence ocular disease. Studies of gut dysbiosis have shown that alterations in the healthy microbiota can predispose the host to immune-mediated inflammatory and neurodegenerative conditions, while oral and ocular surface dysbiosis has been correlated with glaucoma. While developmental exposure to commensal microflora has shown to be necessary for the autoimmune and neurodegenerative responses to elevated intraocular pressure to take place, commensal bacterial products like short-chain fatty acids have regulatory effects protective against glaucoma. SUMMARY Alterations to human microbiotas have been associated with changes in intestinal permeability, gene regulation, immune cell differentiation, and neural functioning, which may predispose the host to glaucoma. Select microbes have been highlighted for their potential contributions to glaucoma disease progression or protection, raising the potential for microbiota-based treatment modalities. Current topical glaucoma treatments may disrupt the ocular surface microbiota, potentially having ramifications on host health. Further study of the relationships between human microbiome and glaucoma is needed.
Collapse
Affiliation(s)
| | - Benjamin Liu
- Department of Ophthalmology, Columbia University Medical Center, New York-Presbyterian Hospital, New York, NY
| | | | | | - Bryan J Winn
- Department of Ophthalmology, Columbia University Medical Center, New York-Presbyterian Hospital, New York, NY
- Ophthalmology Section, Surgical Service, San Francisco Veterans Affairs Medical Center, San Francisco, CA
| |
Collapse
|
41
|
Dinakis E, O'Donnell JA, Marques FZ. The gut-immune axis during hypertension and cardiovascular diseases. Acta Physiol (Oxf) 2024; 240:e14193. [PMID: 38899764 DOI: 10.1111/apha.14193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/04/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
The gut-immune axis is a relatively novel phenomenon that provides mechanistic links between the gut microbiome and the immune system. A growing body of evidence supports it is key in how the gut microbiome contributes to several diseases, including hypertension and cardiovascular diseases (CVDs). Evidence over the past decade supports a causal link of the gut microbiome in hypertension and its complications, including myocardial infarction, atherosclerosis, heart failure, and stroke. Perturbations in gut homeostasis such as dysbiosis (i.e., alterations in gut microbial composition) may trigger immune responses that lead to chronic low-grade inflammation and, ultimately, the development and progression of these conditions. This is unsurprising, as the gut harbors one of the largest numbers of immune cells in the body, yet is a phenomenon not entirely understood in the context of cardiometabolic disorders. In this review, we discuss the role of the gut microbiome, the immune system, and inflammation in the context of hypertension and CVD, and consolidate current evidence of this complex interplay, whilst highlighting gaps in the literature. We focus on diet as one of the major modulators of the gut microbiota, and explain key microbial-derived metabolites (e.g., short-chain fatty acids, trimethylamine N-oxide) as potential mediators of the communication between the gut and peripheral organs such as the heart, arteries, kidneys, and the brain via the immune system. Finally, we explore the dual role of both the gut microbiome and the immune system, and how they work together to not only contribute, but also mitigate hypertension and CVD.
Collapse
Affiliation(s)
- Evany Dinakis
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Joanne A O'Donnell
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, Victoria, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Victorian Heart Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
42
|
Schneider E, O'Riordan KJ, Clarke G, Cryan JF. Feeding gut microbes to nourish the brain: unravelling the diet-microbiota-gut-brain axis. Nat Metab 2024; 6:1454-1478. [PMID: 39174768 DOI: 10.1038/s42255-024-01108-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 07/15/2024] [Indexed: 08/24/2024]
Abstract
The prevalence of brain disorders, including stress-related neuropsychiatric disorders and conditions with cognitive dysfunction, is rising. Poor dietary habits contribute substantially to this accelerating trend. Conversely, healthy dietary intake supports mood and cognitive performance. Recently, the communication between the microorganisms within the gastrointestinal tract and the brain along the gut-brain axis has gained prominence as a potential tractable target to modulate brain health. The composition and function of the gut microbiota is robustly influenced by dietary factors to alter gut-brain signalling. To reflect this interconnection between diet, gut microbiota and brain functioning, we propose that a diet-microbiota-gut-brain axis exists that underpins health and well-being. In this Review, we provide a comprehensive overview of the interplay between diet and gut microbiota composition and function and the implications for cognition and emotional functioning. Important diet-induced effects on the gut microbiota for the development, prevention and maintenance of neuropsychiatric disorders are described. The diet-microbiota-gut-brain axis represents an uncharted frontier for brain health diagnostics and therapeutics across the lifespan.
Collapse
Affiliation(s)
| | | | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
43
|
Zheng B, Wang L, Yi Y, Yin J, Liang A. Design strategies, advances and future perspectives of colon-targeted delivery systems for the treatment of inflammatory bowel disease. Asian J Pharm Sci 2024; 19:100943. [PMID: 39246510 PMCID: PMC11375318 DOI: 10.1016/j.ajps.2024.100943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/02/2024] [Accepted: 05/21/2024] [Indexed: 09/10/2024] Open
Abstract
Inflammatory bowel diseases (IBD) significantly contribute to high mortality globally and negatively affect patients' qualifications of life. The gastrointestinal tract has unique anatomical characteristics and physiological environment limitations. Moreover, certain natural or synthetic anti-inflammatory drugs are associated with poor targeting, low drug accumulation at the lesion site, and other side effects, hindering them from exerting their therapeutic effects. Colon-targeted drug delivery systems represent attractive alternatives as novel carriers for IBD treatment. This review mainly discusses the treatment status of IBD, obstacles to drug delivery, design strategies of colon-targeted delivery systems, and perspectives on the existing complementary therapies. Moreover, based on recent reports, we summarized the therapeutic mechanism of colon-targeted drug delivery. Finally, we addressed the challenges and future directions to facilitate the exploitation of advanced nanomedicine for IBD therapy.
Collapse
Affiliation(s)
- Baoxin Zheng
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Liping Wang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yan Yi
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jun Yin
- School of Traditional Chinese Material, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Aihua Liang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| |
Collapse
|
44
|
Chen YJ, Ho HJ, Tseng CH, Chen YF, Wang ST, Shieh JJ, Wu CY. Short-chain fatty acids ameliorate imiquimod-induced skin thickening and IL-17 levels and alter gut microbiota in mice: a metagenomic association analysis. Sci Rep 2024; 14:17495. [PMID: 39079980 PMCID: PMC11289318 DOI: 10.1038/s41598-024-67325-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
Short-chain fatty acids (SCFAs) have been proposed to have anti-inflammatory effects and improve immune homeostasis. We aimed to examine the effects of SCFAs on skin phenotype, systemic inflammation, and gut microbiota in mice with psoriasis-like inflammation. Imiquimod (IMQ)-treated C57BL/6 mice served as the study model. We conducted a metagenomic association study of IMQ-mice treated with SCFAs or anti-IL-17 antibody using whole-genome shotgun sequencing. The associations among SCFA supplements, skin thickness, circulating inflammatory profiles, and fecal microbiota profiles were investigated. The microbiome study was performed using pipelines for phylogenetic analysis, functional gene analysis, and pathway analysis. In IMQ-treated mice, there were increases in skin thickness and splenic weight, as well as unique fecal microbial profiles. SCFAs ameliorated IMQ-induced skin thickening, splenic weight gain, and serum IL-17F levels, with results that were comparable with those receiving anti-IL-17 treatment. IMQ-treated mice receiving SCFAs had greater microbial diversity than mice treated with IMQ alone. SCFAs and anti-IL17 treatment were associated with alteration of gut microbiota, with increased prevalences of Oscillospiraceae and Lachnopiraceae and decreased prevalences of Muribaculaceae and Bacteroides, which have been predicted to be associated with increased glycan degradation, phenylalanine metabolism, and xylene degradation. SCFAs may mitigate IMQ-induced skin thickening and IL-17F levels and alter fecal microbiota profiles in IMQ-treated mice.
Collapse
Affiliation(s)
- Yi-Ju Chen
- Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan.
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan.
| | - Hsiu J Ho
- Institute of Bioinformatics and Biomedicine, National Yang Ming Chao Tung University, Taipei, Taiwan
| | - Ching-Hung Tseng
- Germark Biotechnology Ltd., No. 21, Keyuan Rd., Situn Dist., Taichung, Taiwan
| | - Yu-Feng Chen
- Institute of Bioinformatics and Biomedicine, National Yang Ming Chao Tung University, Taipei, Taiwan
| | - Sin-Ting Wang
- Institute of Bioinformatics and Biomedicine, National Yang Ming Chao Tung University, Taipei, Taiwan
| | - Jeng-Jer Shieh
- Institute of Biomedicine, National Chung Hsing University, Taichung, Taiwan
| | - Chun-Ying Wu
- Institute of Bioinformatics and Biomedicine, National Yang Ming Chao Tung University, Taipei, Taiwan.
- Division of Translational Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.
- Faculty of Medicine and Graduate Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Department of Public Health and Graduate Institute of Clinical Medical Sciences, China Medical University, Taichung, Taiwan.
| |
Collapse
|
45
|
Munteanu C, Schwartz B. Interactions between Dietary Antioxidants, Dietary Fiber and the Gut Microbiome: Their Putative Role in Inflammation and Cancer. Int J Mol Sci 2024; 25:8250. [PMID: 39125822 PMCID: PMC11311432 DOI: 10.3390/ijms25158250] [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: 06/20/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
The intricate relationship between the gastrointestinal (GI) microbiome and the progression of chronic non-communicable diseases underscores the significance of developing strategies to modulate the GI microbiota for promoting human health. The administration of probiotics and prebiotics represents a good strategy that enhances the population of beneficial bacteria in the intestinal lumen post-consumption, which has a positive impact on human health. In addition, dietary fibers serve as a significant energy source for bacteria inhabiting the cecum and colon. Research articles and reviews sourced from various global databases were systematically analyzed using specific phrases and keywords to investigate these relationships. There is a clear association between dietary fiber intake and improved colon function, gut motility, and reduced colorectal cancer (CRC) risk. Moreover, the state of health is reflected in the reciprocal and bidirectional relationships among food, dietary antioxidants, inflammation, and body composition. They are known for their antioxidant properties and their ability to inhibit angiogenesis, metastasis, and cell proliferation. Additionally, they promote cell survival, modulate immune and inflammatory responses, and inactivate pro-carcinogens. These actions collectively contribute to their role in cancer prevention. In different investigations, antioxidant supplements containing vitamins have been shown to lower the risk of specific cancer types. In contrast, some evidence suggests that taking antioxidant supplements can increase the risk of developing cancer. Ultimately, collaborative efforts among immunologists, clinicians, nutritionists, and dietitians are imperative for designing well-structured nutritional trials to corroborate the clinical efficacy of dietary therapy in managing inflammation and preventing carcinogenesis. This review seeks to explore the interrelationships among dietary antioxidants, dietary fiber, and the gut microbiome, with a particular focus on their potential implications in inflammation and cancer.
Collapse
Affiliation(s)
- Camelia Munteanu
- Department of Plant Culture, Faculty of Agriculture, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| | - Betty Schwartz
- The Institute of Biochemistry, Food Science and Nutrition, The School of Nutritional Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| |
Collapse
|
46
|
Li Y, Wang Y, Li Y, Yan S, Gao X, Li P, Zheng X, Gu Q. Dress me an outfit: advanced probiotics hybrid systems for intelligent IBD therapy. Crit Rev Food Sci Nutr 2024:1-24. [PMID: 39007752 DOI: 10.1080/10408398.2024.2359135] [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: 07/16/2024]
Abstract
Inflammation bowel disease (IBD) has emerged as a public health challenge worldwide; with high incidence and rapid prevalence, it has troubled billions of people and further induced multitudinous systemic complications. Recent decade has witnessed the vigorous application of food-borne probiotics for IBD therapy; however, the complicated and changeable environments of digestive tract have forced probiotics to face multiple in vivo pressures, consequently causing unsatisfied prophylactic or therapeutic efficacy attributed to off-targeted arrival, damaged viability, insufficient colonization efficiency, etc. Fortunately, arisen hybrid technology has provided versatile breakthroughs for the targeted transplantation of probiotics. By ingeniously modifying probiotics to form probiotics hybrid systems (PHS), the biological behaviors of probiotics in vivo could be mediated, the interactions between probiotics with intestinal components can be facilitated, and diverse advanced probiotic-based therapies for IBD challenge can be developed, which attribute to the intelligent response to microenvironment of PHS, and intelligent design of PHS for multiple functions combination. In this review, various PHS were categorized and their intestinal behaviors were elucidated systematically, their therapeutic effects and intrinsic mechanism were further analyzed. Besides, shortages of present PHS and the corresponding solutions have been discussed, based on which the future perspectives of this field have also been proposed. The undeniable fact is that PHS show an incomparable future to bring the next generation of advanced food science.
Collapse
Affiliation(s)
- Yonglu Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Yadi Wang
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Yapeng Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Shihai Yan
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Xin Gao
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Ping Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaodong Zheng
- Department of Food Science and Nutrition; Zhejiang Key Laboratory for Agro-food Processing; Fuli Institute of Food Science; National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou, People's Republic of China
| | - Qing Gu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
47
|
Ji H, Zhong Y, Zhang Z, Chen Y, Zhang Y, Bian S, Yin J, Hu J, Nie S. In vitro digestion and fermentation characteristics of eight kinds of pulses and suggestions for different populations. Food Funct 2024; 15:7314-7332. [PMID: 38898712 DOI: 10.1039/d4fo00551a] [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: 06/21/2024]
Abstract
Pulse-based diets are attracting attention for their potential in combating diet-related non-communicable diseases. However, limited research studies have focused on the digestive and fermentative properties of pulses, which are crucial for exerting benefits. Here, we investigated the in vitro digestibility of starch/protein, along with the fermentation characteristics, of eight pulses and their pastes, including white kidney beans, adzuki beans, cowpeas, broad beans, mung beans, chickpeas, white lentils, and yellow peas. The findings indicated that pulse flours and pastes were low GL food (estimated GL < 10) and had a low degree of protein hydrolysis during simulated gastrointestinal digestion. During in vitro fermentation, pulses flours and pastes decreased the fermentation pH, increased the level of short-chain fatty acids (mainly consisting of valeric acid, followed by acetic acid, propionic acid, butyric acid, isobutyric acid, and isovaleric acid), and positively modulated the microbiota composition over time, specifically reducing the ratio of Firmicutes to Bacteroidetes. In addition, we found that boiling could affect the in vitro digestion and fermentation characteristics of pulses, possibly depending on their intrinsic nutrient characteristics. This research could provide a comprehensive summary of the nutrient content, digestibility, and fermentation of eight pulses and their pastes. Guided by factor analysis, for different individuals' consumption, pulses, cowpeas, broad beans, white lentils, and white kidney beans were preferred for diabetic individuals, yellow peas and white lentils were preferred for intestinal homeostasis disorders, and white lentils, broad beans, white kidney beans, and cowpeas were suitable for obese individuals, in which white lentils were considered healthier and suggested for healthy adults.
Collapse
Affiliation(s)
- Haihua Ji
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Yadong Zhong
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Ziyi Zhang
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Yu Chen
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Yanli Zhang
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Shuigen Bian
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Junyi Yin
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Jielun Hu
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Shaoping Nie
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| |
Collapse
|
48
|
Jama HA, Snelson M, Schutte AE, Muir J, Marques FZ. Recommendations for the Use of Dietary Fiber to Improve Blood Pressure Control. Hypertension 2024; 81:1450-1459. [PMID: 38586958 DOI: 10.1161/hypertensionaha.123.22575] [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: 04/09/2024]
Abstract
According to several international, regional, and national guidelines on hypertension, lifestyle interventions are the first-line treatment to lower blood pressure (BP). Although diet is one of the major lifestyle modifications described in hypertension guidelines, dietary fiber is not specified. Suboptimal intake of foods high in fiber, such as in Westernized diets, is a major contributing factor to mortality and morbidity of noncommunicable diseases due to higher BP and cardiovascular disease. In this review, we address this deficiency by examining and advocating for the incorporation of dietary fiber as a key lifestyle modification to manage elevated BP. We explain what dietary fiber is, review the existing literature that supports its use to lower BP and prevent cardiovascular disease, describe the mechanisms involved, propose evidence-based target levels of fiber intake, provide examples of how patients can achieve the recommended targets, and discuss outstanding questions in the field. According to the evidence reviewed here, the minimum daily dietary fiber for adults with hypertension should be >28 g/day for women and >38 g/day for men, with each extra 5 g/day estimated to reduce systolic BP by 2.8 mm Hg and diastolic BP by 2.1 mm Hg. This would support a healthy gut microbiota and the production of gut microbiota-derived metabolites called short-chain fatty acids that lower BP. Awareness about dietary fiber targets and how to achieve them will guide medical teams on better educating patients and empowering them to increase their fiber intake and, as a result, lower their BP and cardiovascular disease risk.
Collapse
Affiliation(s)
- Hamdi A Jama
- Hypertension Research Laboratory, School of Biological Sciences (H.A.J., M.S., F.Z.M.), Monash University, Melbourne, VIC, Australia
| | - Matthew Snelson
- Hypertension Research Laboratory, School of Biological Sciences (H.A.J., M.S., F.Z.M.), Monash University, Melbourne, VIC, Australia
- Victorian Heart Institute (M.S., F.Z.M.), Monash University, Melbourne, VIC, Australia
| | - Aletta E Schutte
- School of Population Health, University of New South Wales, Sydney, Australia (A.E.S.)
- George Institute for Global Health, Sydney, NSW, Australia (A.E.S.)
- Hypertension in Africa Research Team, MRC Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa (A.E.S.)
| | - Jane Muir
- Department of Gastroenterology, School of Translational Medicine (J.M.), Monash University, Melbourne, VIC, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences (H.A.J., M.S., F.Z.M.), Monash University, Melbourne, VIC, Australia
- Victorian Heart Institute (M.S., F.Z.M.), Monash University, Melbourne, VIC, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (F.Z.M.)
| |
Collapse
|
49
|
Sasaki M, Suaini NHA, Afghani J, Heye KN, O'Mahony L, Venter C, Lauener R, Frei R, Roduit C. Systematic review of the association between short-chain fatty acids and allergic diseases. Allergy 2024; 79:1789-1811. [PMID: 38391245 DOI: 10.1111/all.16065] [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: 09/26/2023] [Revised: 01/23/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024]
Abstract
We performed a systematic review to investigate the current evidence on the association between allergic diseases and short chain fatty acids (SCFAs), which are microbially produced and suggested as one mechanism on how gut microbiome affects the risk of allergic diseases. Medline, Embase and Web of Science were searched from data inception until September 2022. We identified 37 papers, of which 17 investigated prenatal or early childhood SCFAs and the development of allergic diseases in childhood, and 20 assessed SCFAs in patients with pre-existing allergic diseases. Study design, study populations, outcome definition, analysis method and reporting of the results varied between papers. Overall, there was some evidence showing that the three main SCFAs (acetate, propionate and butyrate) in the first few years of life had a protective effect against allergic diseases, especially for atopic dermatitis, wheeze or asthma and IgE-mediated food allergy in childhood. The association between each SCFA and allergic disease appeared to be different by disease and the age of assessment. Further research that can determine the potentially timing specific effect of each SCFA will be useful to investigate how SCFAs can be used in treatment or in prevention against allergic diseases.
Collapse
Affiliation(s)
- Mari Sasaki
- University Children's Hospital Zürich, Zürich, Switzerland
- Division of Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, University of Bern, Bern, Switzerland
| | - Noor H A Suaini
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jamie Afghani
- Environmental Medicine Faculty of Medicine, University of Augsburg, Augsburg, Germany
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
- Institute of Environmental Medicine, Environmental Health Centre, Helmholtz Munich - German Research Centre for Environmental Health (GmbH), Neuherberg, Germany
| | - Kristina N Heye
- Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
| | - Liam O'Mahony
- Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland
- APC Microbiome Ireland, University College Cork, National University of Ireland, Cork, Ireland
- School of Microbiology, University College Cork, National University of Ireland, Cork, Ireland
| | - Carina Venter
- Pediatric Allergy and Immunology, University of Colorado/Childrens Hospital Colorado, Aurora, Colorado, USA
| | - Roger Lauener
- Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
- Christine Kühne Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Remo Frei
- Division of Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, University of Bern, Bern, Switzerland
- Christine Kühne Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Caroline Roduit
- University Children's Hospital Zürich, Zürich, Switzerland
- Division of Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, University of Bern, Bern, Switzerland
- Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
- Christine Kühne Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| |
Collapse
|
50
|
Ozturk O, Celebi G, Duman UG, Kupcuk E, Uyanik M, Sertoglu E. Short-chain fatty acid levels in stools of patients with inflammatory bowel disease are lower than those in healthy subjects. Eur J Gastroenterol Hepatol 2024; 36:890-896. [PMID: 38829943 DOI: 10.1097/meg.0000000000002789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
OBJECTIVE Short-chain fatty acids (SCFAs) are produced when the microbiota in the large intestine cause fermentation of dietary carbohydrates and fibers. These fatty acids constitute the primary energy source of colon mucosa cells and have a protective effect in patients suffering from inflammatory bowel disease (IBD). This study aimed to compare the SCFA levels in the stools of patients with IBD and healthy controls. METHOD Healthy controls and patients with IBD aged 18 and over were included in the study. Stool samples from all patients and healthy controls were collected, and stool acetic acid, propionic acid, and butyric acid levels were measured using a gas chromatography-mass spectrometry measurement method. RESULTS In this study, 64 participants were divided into two groups: 34 were in IBD (Crohn disease and ulcerative colitis) and 30 were in healthy control group. When fecal SCFA concentrations of IBD and healthy control groups were compared, a statistically significant difference was observed between them. When the fecal SCFA concentrations of Crohn's disease and ulcerative colitis patients in the IBD group were compared, however, no statistically significant difference was observed between them. Furthermore, when the participants' diet type (carbohydrate-based, vegetable-protein-based and mixed diet) and the number of meals were compared with fecal SCFA concentrations, no statistically significant difference was observed between them. CONCLUSION In general, fecal SCFA levels in patients with IBD were lower than those in healthy controls. Moreover, diet type and the number of meals had no effect on stool SCFA levels in patients with IBD and healthy individuals.
Collapse
Affiliation(s)
| | - Gurkan Celebi
- Department of Gastroenterology, Gulhane School of Medicine, University of Health Sciences, Ankara
| | | | | | - Metin Uyanik
- Department of Biochemistry, Çorlu State Hospital, Tekirdag, Turkey
| | | |
Collapse
|