1
|
Zhang S, Niu H, Zhu J. Personalized nutrition studies of human gut microbiome-polyphenol interactions utilizing continuous multistaged in vitro fermentation models-a narrative review. Nutr Res 2025; 135:101-127. [PMID: 39999639 DOI: 10.1016/j.nutres.2025.01.011] [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: 08/22/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025]
Abstract
The gut microbiota, a complex community of microorganisms primarily inhabiting the human large intestine, plays a crucial role in human health. Gut dysbiosis, characterized by an imbalance in gut bacterial populations, has been increasingly recognized as a significant factor in the pathogenesis of metabolic diseases such as type 2 diabetes, inflammatory bowel disease, and colorectal cancer. Polyphenols are critical modulators of gut microbial composition and metabolism. However, the extent of polyphenol-induced modulation of the gut microbiome remains largely unexplored. In vitro models offer a convenient and ethical alternative to in vivo studies for investigating nutrient-gut microbiome interactions, facilitating easy sampling and controlled experimental conditions. Among these, continuous multistaged in vitro fermentation models, which simulate different sections of the human gastrointestinal tract (e.g., proximal colon, transverse colon, and distal colon), provide a more accurate representation of the human gut environment compared to single-batch fermentation. Various configurations of these multistaged models have been developed and widely employed in studies examining the effects of polyphenols on the gut microbiome. This review aims to summarize the different configurations of multistaged in vitro fermentation models and recent advancements in their development, highlight key aspects of experimental design, outline commonly used analytical workflows with complementary analyses, and review the restorative effects of polyphenol interventions on dysregulated gut microbiota.
Collapse
Affiliation(s)
- Shiqi Zhang
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA
| | - Hanmeng Niu
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA
| | - Jiangjiang Zhu
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
2
|
Gülşan EE, Nowshad F, Leigh MD, Crott JW, Park H, Martin G, Safe S, Chapkin RS, Jayaraman A, Lee K. A Chalcone Synthase-like Bacterial Protein Catalyzes Heterocyclic C-Ring Cleavage of Naringenin to Alter Bioactivity Against Nuclear Receptors in Colonic Epithelial Cells. Metabolites 2025; 15:146. [PMID: 40137111 PMCID: PMC11943482 DOI: 10.3390/metabo15030146] [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: 01/07/2025] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Gut microbial metabolism of dietary flavonoids leads to a diverse array of bioactive products that are closely associated with human health. Combining enzyme promiscuity prediction, metabolomics, and in vitro model systems, we identified a chalcone-synthase-like bacterial polyketide synthase that can initiate the metabolism of naringenin by catalyzing the C-ring cleavage. This was validated using a mutant strain of the model organism Bacillus subtilis (ATCC 23857). Our prediction-validation methodology could be used to systematically characterize the products of gut bacterial flavonoid metabolism and identify the responsible enzymes and species. In vitro experiments with Caco-2 cells revealed that naringenin and its bacterial metabolites differentially engage the aryl hydrocarbon receptor (AhR) and orphan nuclear receptor 4A (NR4A). These results suggest that metabolism by gut bacterial species could directly impact the profile of bioactive flavonoids and influence inflammatory responses in the intestine. These results are significant for understanding gut-microbiota-dependent physiological effects of dietary flavonoids.
Collapse
Affiliation(s)
- Ebru Ece Gülşan
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA 02155, USA;
| | - Farrhin Nowshad
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX 77843, USA;
| | - Meredith Davis Leigh
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA;
| | - Jimmy Walter Crott
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA;
| | - Hyejin Park
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (H.P.); (G.M.); (S.S.)
| | - Greg Martin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (H.P.); (G.M.); (S.S.)
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (H.P.); (G.M.); (S.S.)
| | - Robert S. Chapkin
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA;
| | - Arul Jayaraman
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX 77843, USA;
| | - Kyongbum Lee
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA 02155, USA;
| |
Collapse
|
3
|
Ren P, Wei B, Qin W, Tang Q, Wang Y, Xue C. Impact of astaxanthin on the capacity of gut microbiota to produce tryptophan catabolites. Food Funct 2025; 16:524-538. [PMID: 39688008 DOI: 10.1039/d4fo04890c] [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: 12/18/2024]
Abstract
This study utilized in vitro colonic fermentation to examine the impact of astaxanthin on the microbial catabolism of tryptophan. Astaxanthin significantly altered the gut microbiota and raised the tryptophan catabolism metabolite levels in an in vitro human colonic fermentation system. To eliminate the influence of substrate availability, we conducted in vitro colonic fermentation of the gut microbiota of astaxanthin-domesticated mice. We observed that the capacity of astaxanthin-domesticated gut microbiota to catalyze the conversion of tryptophan to indole and derivatives was considerably augmented. Astaxanthin significantly increased the relative abundance of Akkermansia, Ruminococcus, Bacteroides and Lactobacillus and elevated the levels of indole-3-lactic acid and indole-3-propionic acid. These results demonstrated that astaxanthin regulates tryptophan metabolism by modifying gut microbiota and increasing the levels of indole metabolites, such as indole-3-lactic acid and indole-3-propionic acid. This study provides insights into the physiological activity of astaxanthin and sheds light on the potential for enhancing tryptophan metabolism through dietary manipulation of the gut microbiota.
Collapse
Affiliation(s)
- Pengfei Ren
- State Key Laboratory of Marine Food Processing &Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, China.
| | - Biqian Wei
- State Key Laboratory of Marine Food Processing &Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, China.
| | - Wanting Qin
- State Key Laboratory of Marine Food Processing &Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, China.
| | - Qingjuan Tang
- State Key Laboratory of Marine Food Processing &Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, China.
| | - Yuming Wang
- State Key Laboratory of Marine Food Processing &Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, China.
| | - Changhu Xue
- State Key Laboratory of Marine Food Processing &Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, China.
| |
Collapse
|
4
|
Dell'Olio A, Rubert J, Capozzi V, Tonezzer M, Betta E, Fogliano V, Biasioli F. Non-invasive VOCs detection to monitor the gut microbiota metabolism in-vitro. Sci Rep 2024; 14:15842. [PMID: 38982163 PMCID: PMC11233675 DOI: 10.1038/s41598-024-66303-7] [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] [Received: 03/01/2024] [Accepted: 07/01/2024] [Indexed: 07/11/2024] Open
Abstract
This work implemented a non-invasive volatile organic compounds (VOCs) monitoring approach to study how food components are metabolised by the gut microbiota in-vitro. The fermentability of a model food matrix rich in dietary fibre (oat bran), and a pure prebiotic (inulin), added to a minimal gut medium was compared by looking at global changes in the volatilome. The substrates were incubated with a stabilised human faecal inoculum over a 24-h period, and VOCs were monitored without interfering with biological processes. The fermentation was performed in nitrogen-filled vials, with controlled temperature, and tracked by automated headspace-solid-phase microextraction coupled with gas chromatography-mass spectrometry. To understand the molecular patterns over time, we applied a multivariate longitudinal statistical framework: repeated measurements-ANOVA simultaneous component analysis. The methodology was able to discriminate the studied groups by looking at VOCs temporal profiles. The volatilome showed a time-dependency that was more distinct after 12 h. Short to medium-chain fatty acids showed increased peak intensities, mainly for oat bran and for inulin, but with different kinetics. At the same time, alcohols, aldehydes, and esters showed distinct trends with discriminatory power. The proposed approach can be applied to study the intertwined pathways of gut microbiota food components interaction in-vitro.
Collapse
Affiliation(s)
- Andrea Dell'Olio
- Food Quality and Design, Wageningen University & Research, 6708 WG, Wageningen, Netherlands
- Reserach and Innovation Centre, Fondazione Edmund Mach, 39098, San Michele All'Adige, Italy
| | - Josep Rubert
- Food Quality and Design, Wageningen University & Research, 6708 WG, Wageningen, Netherlands
| | - Vittorio Capozzi
- Institute of Food Production Sciences, National Research Council, 71121, Foggia, Italy
| | - Matteo Tonezzer
- Reserach and Innovation Centre, Fondazione Edmund Mach, 39098, San Michele All'Adige, Italy
- Department of Chemical and Geological Sciences, University of Cagliari, 09042, Monserrato , Italy
| | - Emanuela Betta
- Reserach and Innovation Centre, Fondazione Edmund Mach, 39098, San Michele All'Adige, Italy
| | - Vincenzo Fogliano
- Food Quality and Design, Wageningen University & Research, 6708 WG, Wageningen, Netherlands
| | - Franco Biasioli
- Reserach and Innovation Centre, Fondazione Edmund Mach, 39098, San Michele All'Adige, Italy.
| |
Collapse
|
5
|
Han YZ, Zheng HJ, Du BX, Zhang Y, Zhu XY, Li J, Wang YX, Liu WJ. Role of Gut Microbiota, Immune Imbalance, and Allostatic Load in the Occurrence and Development of Diabetic Kidney Disease. J Diabetes Res 2023; 2023:8871677. [PMID: 38094870 PMCID: PMC10719010 DOI: 10.1155/2023/8871677] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 11/15/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Diabetic kidney disease (DKD) is a prevailing complication arising from diabetes mellitus. Unfortunately, there are no trustworthy and efficacious treatment modalities currently available. In recent times, compelling evidence has emerged regarding the intricate correlation between the kidney and the gut microbiota, which is considered the largest immune organ within the human physique. Various investigations have demonstrated that the perturbation of the gut microbiota and its associated metabolites potentially underlie the etiology and progression of DKD. This phenomenon may transpire through perturbation of both the innate and the adaptive immunity, leading to a burdensome allostatic load on the body and ultimately culminating in the development of DKD. Within this literature review, we aim to delve into the intricate interplay between the gut microbiota, its metabolites, and the immune system in the context of DKD. Furthermore, we strive to explore and elucidate potential chemical interventions that could hold promise for the treatment of DKD, thereby offering invaluable insights and directions for future research endeavors.
Collapse
Affiliation(s)
- Yi Zhen Han
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Juan Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Bo Xuan Du
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xing Yu Zhu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Li
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Yao Xian Wang
- Beijing University of Chinese Medicine, Beijing, China
| | - Wei Jing Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
6
|
Huang Z, Boekhorst J, Fogliano V, Capuano E, Wells JM. Impact of High-Fiber or High-Protein Diet on the Capacity of Human Gut Microbiota To Produce Tryptophan Catabolites. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:6956-6966. [PMID: 37126824 PMCID: PMC10176579 DOI: 10.1021/acs.jafc.2c08953] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
This study investigated the effect of high-fiber-low-protein (HF) and high-protein-low-fiber (HP) diets on microbial catabolism of tryptophan in the proximal colon (PC) and distal colon(DC) compartments of the Simulator of the Human Intestinal Microbial Ecosystem. The microbiota in PC and DC was dominated by Bacteroidetes and Firmicutes, in which Bacteroidetes were more abundant in DC (∼60% versus 50%) and Firmicutes were more abundant in PC (∼40% versus 25%). Most of the tryptophan catabolites were determined at a higher concentration in PC samples than in DC samples, but the overall concentration of tryptophan catabolites was over 10-fold higher in DC samples than that in PC samples. Interestingly, indole-3-propionic acid and oxindole were only identified in DC samples. A two-week dietary intervention by the HF diet enriched the abundance of Firmicutes in PC, whereas the HP diet enriched the abundance of Proteobacteria. Compared to the HP diet, the HF diet favored the microbial production of indole-3-acetic acid, indole-3-lactic acid, indole-3-aldehyde, and indole-3-propionic acid in both PC and DC compartments. To conclude, these findings increase the understanding of the effect of diets on the microbial production of tryptophan catabolites in the colon.
Collapse
Affiliation(s)
- Zhan Huang
- Food Quality and Design Group, Department of Agrotechnology and Food Sciences, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
- Host-Microbe Interactomics Group, Department of Animal Sciences, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
| | - Jos Boekhorst
- Host-Microbe Interactomics Group, Department of Animal Sciences, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
| | - Vincenzo Fogliano
- Food Quality and Design Group, Department of Agrotechnology and Food Sciences, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
| | - Edoardo Capuano
- Food Quality and Design Group, Department of Agrotechnology and Food Sciences, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
| | - Jerry M Wells
- Host-Microbe Interactomics Group, Department of Animal Sciences, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
| |
Collapse
|
7
|
Pinto CJG, Ávila-Gálvez MÁ, Lian Y, Moura-Alves P, Nunes Dos Santos C. Targeting the aryl hydrocarbon receptor by gut phenolic metabolites: A strategy towards gut inflammation. Redox Biol 2023; 61:102622. [PMID: 36812782 PMCID: PMC9958510 DOI: 10.1016/j.redox.2023.102622] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
The Aryl Hydrocarbon Receptor (AHR) is a ligand-dependent transcription factor able to control complex transcriptional processes in several cell types, which has been correlated with various diseases, including inflammatory bowel diseases (IBD). Numerous studies have described different compounds as ligands of this receptor, like xenobiotics, natural compounds, and several host-derived metabolites. Dietary (poly)phenols have been studied regarding their pleiotropic activities (e.g., neuroprotective and anti-inflammatory), but their AHR modulatory capabilities have also been considered. However, dietary (poly)phenols are submitted to extensive metabolism in the gut (e.g., gut microbiota). Thus, the resulting gut phenolic metabolites could be key players modulating AHR since they are the ones that reach the cells and may exert effects on the AHR throughout the gut and other organs. This review aims at a comprehensive search for the most abundant gut phenolic metabolites detected and quantified in humans to understand how many have been described as AHR modulators and what could be their impact on inflammatory gut processes. Even though several phenolic compounds have been studied regarding their anti-inflammatory capacities, only 1 gut phenolic metabolite, described as AHR modulator, has been evaluated on intestinal inflammatory models. Searching for AHR ligands could be a novel strategy against IBD.
Collapse
Affiliation(s)
- Catarina J G Pinto
- iNOVA4Health, NOVA Medical School
- Faculdade de Ciências Médicas, NMS
- FCM, Universidade Nova de Lisboa, Lisboa, Portugal; IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal; I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - María Ángeles Ávila-Gálvez
- iNOVA4Health, NOVA Medical School
- Faculdade de Ciências Médicas, NMS
- FCM, Universidade Nova de Lisboa, Lisboa, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, Portugal
| | - Yilong Lian
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, OX3 7DQ, Oxford, United Kingdom
| | - Pedro Moura-Alves
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal; I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal; Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, OX3 7DQ, Oxford, United Kingdom.
| | - Cláudia Nunes Dos Santos
- iNOVA4Health, NOVA Medical School
- Faculdade de Ciências Médicas, NMS
- FCM, Universidade Nova de Lisboa, Lisboa, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, Portugal.
| |
Collapse
|
8
|
Liu C, Gan RY, Chen D, Zheng L, Ng SB, Rietjens IMCM. Gut microbiota-mediated metabolism of green tea catechins and the biological consequences: An updated review. Crit Rev Food Sci Nutr 2023; 64:7067-7084. [PMID: 38975869 DOI: 10.1080/10408398.2023.2180478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Multiple beneficial effects have been attributed to green tea catechins (GTCs). However, the bioavailability of GTCs is generally low, with only a small portion directly absorbed in the small intestine. The majority of ingested GTCs reaches the large intestinal lumen, and are extensively degraded via biotransformation by gut microbiota, forming many low-molecular-weight metabolites such as phenyl-γ-valerolactones, phenolic acids, butyrate, and acetate. This process not only improves the overall bioavailability of GTC-derived metabolites but also enriches the biological activities of GTCs. Therefore, the intra- and inter-individual differences in human gut microbiota as well as the resulting biological contribution of microbial metabolites are crucial for the ultimate health benefits. In this review, the microbial degradation of major GTCs was characterized and an overview of the in vitro models used for GTC metabolism was summarized. The intra- and inter-individual differences of human gut microbiota composition and the resulting divergence in the metabolic patterns of GTCs were highlighted. Moreover, the potential beneficial effects of GTCs and their gut microbial metabolites were also discussed. Overall, the microbial metabolites of GTCs with higher bioavailability and bioactive potency are key factors for the observed beneficial effects of GTCs and green tea consumption.
Collapse
Affiliation(s)
- Chen Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China
- Division of Toxicology, Wageningen University and Research, Wageningen, the Netherlands
- Tea Refining and Innovation Key Laboratory of Sichuan Province, College of Horticulture, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Ren-You Gan
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Liang Zheng
- Division of Toxicology, Wageningen University and Research, Wageningen, the Netherlands
| | - Siew Bee Ng
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University and Research, Wageningen, the Netherlands
| |
Collapse
|
9
|
Duque-Soto C, Quintriqueo-Cid A, Rueda-Robles A, Robert P, Borrás-Linares I, Lozano-Sánchez J. Evaluation of Different Advanced Approaches to Simulation of Dynamic In Vitro Digestion of Polyphenols from Different Food Matrices-A Systematic Review. Antioxidants (Basel) 2022; 12:101. [PMID: 36670962 PMCID: PMC9854833 DOI: 10.3390/antiox12010101] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Phenolic compounds have become interesting bioactive antioxidant compounds with implications for obesity, cancer and inflammatory gastrointestinal pathologies. As the influence of digestion and gut microbiota on antioxidant behavior is yet to be completely elucidated, and due to limitations associated to in vivo studies, dynamic in vitro gastrointestinal models have been promoted. A systematic review was conducted of different databases (PubMed, Web of Science and Scopus) following PRISMA guidelines to assess different dynamic digestion models and assay protocols used for phenolic compound research regarding bioaccesibility and interaction with colonic microbiota. Of 284 records identified, those including dynamic multicompartmental digestion models for the study of phenolic compound bioaccesibility, bioactivity and the effects of microbiota were included, with 57 studies meeting the inclusion criteria. Different conditions and experimental configurations as well as administered doses, sample treatments and microbiological assays of dynamic digestion studies on polyphenols were recorded and compared to establish their relevance for the dynamic in vitro digestion of phenolic compounds. While similarities were observed in certain experimental areas, a high variability was found in others, such as administered doses. A description of considerations on the study of the digestion of phenolic compounds is proposed to enhance comparability in research.
Collapse
Affiliation(s)
- Carmen Duque-Soto
- Department of Food Science and Nutrition, Faculty of Farmacy, University of Granada, 18071 Granada, Spain
| | - Alejandra Quintriqueo-Cid
- Department of Food Science and Nutrition, Faculty of Farmacy, University of Granada, 18071 Granada, Spain
- Departamento de Ciencia de los Alimentos y Tecnología Química, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 8380492, Chile
| | - Ascensión Rueda-Robles
- Department of Food Science and Nutrition, Faculty of Farmacy, University of Granada, 18071 Granada, Spain
| | - Paz Robert
- Departamento de Ciencia de los Alimentos y Tecnología Química, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 8380492, Chile
| | - Isabel Borrás-Linares
- Department of Analytical Chemistry, Faculty of Science, University of Granada, 18071 Granada, Spain
| | - Jesús Lozano-Sánchez
- Department of Food Science and Nutrition, Faculty of Farmacy, University of Granada, 18071 Granada, Spain
| |
Collapse
|
10
|
Polyphenols as Drivers of a Homeostatic Gut Microecology and Immuno-Metabolic Traits of Akkermansia muciniphila: From Mouse to Man. Int J Mol Sci 2022; 24:ijms24010045. [PMID: 36613488 PMCID: PMC9820369 DOI: 10.3390/ijms24010045] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Akkermansia muciniphila is a mucosal symbiont considered a gut microbial marker in healthy individuals, as its relative abundance is significantly reduced in subjects with gut inflammation and metabolic disturbances. Dietary polyphenols can distinctly stimulate the relative abundance of A. muciniphila, contributing to the attenuation of several diseases, including obesity, type 2 diabetes, inflammatory bowel diseases, and liver damage. However, mechanistic insight into how polyphenols stimulate A. muciniphila or its activity is limited. This review focuses on dietary interventions in rodents and humans and in vitro studies using different phenolic classes. We provide critical insights with respect to potential mechanisms explaining the effects of polyphenols affecting A. muciniphila. Anthocyanins, flavan-3-ols, flavonols, flavanones, stilbenes, and phenolic acids are shown to increase relative A. muciniphila levels in vivo, whereas lignans exert the opposite effect. Clinical trials show consistent findings, and high intervariability relying on the gut microbiota composition at the baseline and the presence of multiple polyphenol degraders appear to be cardinal determinants in inducing A. muciniphila and associated benefits by polyphenol intake. Polyphenols signal to the AhR receptor and impact the relative abundance of A. muciniphila in a direct and indirect fashion, resulting in the restoration of intestinal epithelial integrity and homeostatic crosstalk with the gut microbiota by affecting IL-22 production. Moreover, recent evidence suggests that A. muciniphila participates in the initial hydrolysis of some polyphenols but does not participate in their complete metabolism. In conclusion, the consumption of polyphenol-rich foods targeting A. muciniphila as a pivotal intermediary represents a promising precision nutritional therapy to prevent and attenuate metabolic and inflammatory diseases.
Collapse
|
11
|
Zhang Z, Wang J, Lin Y, Chen J, Liu J, Zhang X. Nutritional activities of luteolin in obesity and associated metabolic diseases: an eye on adipose tissues. Crit Rev Food Sci Nutr 2022; 64:4016-4030. [PMID: 36300856 DOI: 10.1080/10408398.2022.2138257] [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: 11/03/2022]
Abstract
Obesity is characterized by excessive body fat accumulation and is a high-risk factor for metabolic comorbidities, including type 2 diabetes, nonalcoholic fatty liver disease, and cardiovascular disease. In lean individuals, adipose tissue (AT) is not only an important regulatory organ for energy storage and metabolism, but also an indispensable immune and endocrine organ. The sustained energy imbalance induces adipocyte hypotrophy and hyperplasia as well as AT remodeling, accompanied by chronic low-grade inflammation and adipocytes dysfunction in AT, ultimately leading to systemic insulin resistance and ectopic lipid deposition. Luteolin is a natural flavonoid widely distributed in fruits and vegetables and possesses multifold biological activities, such as antioxidant, anticancer, and anti-inflammatory activities. Diet supplementation of this flavonoid has been reported to inhibit AT lipogenesis and inflammation as well as the ectopic lipid deposition, increase AT thermogenesis and systemic energy expenditure, and finally improve obesity and associated metabolic diseases. The purpose of this review is to reveal the nutritional activities of luteolin in obesity and its complications with emphasis on its action on AT energy metabolism, immunoregulation, and endocrine intervention.
Collapse
Affiliation(s)
- Zhixin Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Jiahui Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Yan Lin
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Juan Chen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Jian Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
- Engineering Research Center of Bioprocess, Ministry of Education, Hefei University of Technology, Hefei, Anhui, China
| | - Xian Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| |
Collapse
|
12
|
Liu TH, Xie T, Bai ZY, Liang QE, Xie PC, Xue YZ, Xiao Y, Chen LG. The Important Role of TaohongSiwu Decoction in Gut Microbial Modulation in Response to High-Salt Diet-Induced Hypertensive Mice. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221118199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
TaohongSiwu decoction (THSWD), a traditional Chinese recipe, has been widely used to treat hypertension since ancient times. However, the mechanisms of its action are still unclear. Herein, we aimed to explore the gut microbial activity of THSWD in high-salt diet-induced hypertensive mice. Eight percent high-salt (NaCl) diet was used to induce hypertension for 4 weeks in a mouse model. Meanwhile, THSWD was used to intervene in the high-salt diet-induced mice, and the efficacy was evaluated by different parameters. Here, we found that THSWD significantly restored blood pressure compared with the model group. Moreover, THSWD effectively protected endothelial function by significantly upregulating the level of nitric oxide (NO) and downregulating the level of endothelin-1 (ET-1), angiotensin I (AngI), and vascular endothelial growth factor (VEGF) in serum compared with the model group. Notably, THSWD significantly upregulated the relative abundance of Dubosiella and downregulated that of Cyanobium_PCC-6307 and DNF00809 at the genus level compared with the model group. The results of PCA and microbial distance calculation further exhibited that THSWD treatment resulted in significant regulation of the microbial community. Furthermore, compared with the model group, THSWD increased the level of vitamin k2 (VK2) in serum. These findings indicate that THSWD could protect blood pressure and endothelial function by regulating gut microbiota and promoting microbial metabolite VK2. These results show the important role of THSWD in regulating the gut microbiota in response to high-salt diet-induced mice.
Collapse
Affiliation(s)
- Tian-hao Liu
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, China
- College of Chinese Medicine, Jinan University, Guangzhou, China
| | - Ting Xie
- College of Chinese Medicine, Jinan University, Guangzhou, China
| | - Zhen-yu Bai
- College of Chinese Medicine, Jinan University, Guangzhou, China
| | - Qiu-er Liang
- College of Chinese Medicine, Jinan University, Guangzhou, China
| | - Peng-cheng Xie
- College of Chinese Medicine, Jinan University, Guangzhou, China
| | - Yu-zheng Xue
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Ya Xiao
- College of Chinese Medicine, Jinan University, Guangzhou, China
| | - Li-guo Chen
- College of Chinese Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
13
|
Distinct effects of fiber and colon segment on microbiota-derived indoles and short-chain fatty acids. Food Chem 2022; 398:133801. [DOI: 10.1016/j.foodchem.2022.133801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 07/02/2022] [Accepted: 07/25/2022] [Indexed: 01/04/2023]
|
14
|
Koper J, Troise AD, Loonen LMP, Vitaglione P, Capuano E, Fogliano V, Wells JM. Tryptophan Supplementation Increases the Production of Microbial-Derived AhR Agonists in an In Vitro Simulator of Intestinal Microbial Ecosystem. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:3958-3968. [PMID: 35344652 PMCID: PMC8991005 DOI: 10.1021/acs.jafc.1c04145] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The aryl hydrocarbon receptor (AhR) plays an important role in intestinal homeostasis, and some microbial metabolites of tryptophan are known AhR agonists. In this study, we assessed the impact of tryptophan supplementation on the formation of tryptophan metabolites, AhR activation, and microbiota composition in the simulator of the human intestinal microbial ecosystem (SHIME). AhR activation, microbial composition, and tryptophan metabolites were compared during high tryptophan supplementation (4 g/L tryptophan), control, and wash-out periods. During tryptophan supplementation, the concentration of several tryptophan metabolites was increased compared to the control and wash-out period, but AhR activation by fermenter supernatant was significantly decreased. This was due to the higher levels of tryptophan, which was found to be an antagonist of AhR signaling. Tryptophan supplementation induced most microbial changes in the transverse colon including increased relative abundance of lactobacillus. We conclude that tryptophan supplementation leads to increased formation of AhR agonists in the colon.
Collapse
Affiliation(s)
- Jonna
EB Koper
- Department
of Agrotechnology & Food Sciences, Wageningen
University, Wageningen 6708 WE, The Netherlands
- Department
of Animal Sciences, Wageningen University, Wageningen 6708 WD, The Netherlands
| | - Antonio Dario Troise
- Department
of Food Science, University of Naples “Federico
II”, Parco
Gussone 80055, Italy
| | - Linda MP Loonen
- Department
of Animal Sciences, Wageningen University, Wageningen 6708 WD, The Netherlands
| | - Paola Vitaglione
- Department
of Agricultural Sciences, University of
Naples “Federico II”, Parco Gussone 80055, Italy
| | - Edoardo Capuano
- Department
of Agrotechnology & Food Sciences, Wageningen
University, Wageningen 6708 WE, The Netherlands
| | - Vincenzo Fogliano
- Department
of Agrotechnology & Food Sciences, Wageningen
University, Wageningen 6708 WE, The Netherlands
| | - Jerry M Wells
- Department
of Animal Sciences, Wageningen University, Wageningen 6708 WD, The Netherlands
| |
Collapse
|
15
|
High Throughput Analysis Reveals Changes in Gut Microbiota and Specific Fecal Metabolomic Signature in Hematopoietic Stem Cell Transplant Patients. Microorganisms 2021; 9:microorganisms9091845. [PMID: 34576740 PMCID: PMC8469814 DOI: 10.3390/microorganisms9091845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/02/2021] [Accepted: 08/23/2021] [Indexed: 12/29/2022] Open
Abstract
There is mounting evidence for the emerging role of gut microbiota (GM) and its metabolites in profoundly impacting allogenic hematopoietic stem cell transplantation (allo-HSCT) and its subsequent complications, mainly infections and graft versus host-disease (GvHD). The present study was performed in order to investigate changes in GM composition and fecal metabolic signature between transplant patients (n = 15) and healthy controls (n = 18). The intestinal microbiota was characterized by NGS and gas chromatography-mass spectrometry was employed to perform untargeted analysis of fecal metabolites. We found lower relative abundances of Actinobacteria, Firmicutes, and Bacteroidetes and a higher abundance of Proteobacteria phylum after allo-HSCT. Particularly, the GvHD microbiota was characterized by a lower relative abundance of the short-chain fatty acid-producing bacteria, namely, the Feacalibacterium, Akkermansia, and Veillonella genera and the Lachnospiraceae family, and an enrichment in multidrug-resistant bacteria belonging to Escherichia, Shigella, and Bacteroides. Moreover, network analysis showed that GvHD was linked to a higher number of positive interactions of Blautia and a significant mutual-exclusion rate of Citrobacter. The fecal metabolome was dominated by lipids in the transplant group when compared with the healthy individuals (p < 0.05). Overall, 76 metabolites were significantly altered within transplant recipients, of which 24 were selected as potential biomarkers. Furthermore, the most notable altered metabolic pathways included the TCA cycle; butanoate, propanoate, and pyruvate metabolisms; steroid biosynthesis; and glycolysis/gluconeogenesis. Specific biomarkers and altered metabolic pathways were correlated to GvHD onset. Our results showed significant shifts in gut microbiota structure and fecal metabolites characterizing allo-HSCT.
Collapse
|
16
|
Bucciantini M, Leri M, Nardiello P, Casamenti F, Stefani M. Olive Polyphenols: Antioxidant and Anti-Inflammatory Properties. Antioxidants (Basel) 2021; 10:antiox10071044. [PMID: 34209636 PMCID: PMC8300823 DOI: 10.3390/antiox10071044] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress and inflammation triggered by increased oxidative stress are the cause of many chronic diseases. The lack of anti-inflammatory drugs without side-effects has stimulated the search for new active substances. Plant-derived compounds provide new potential anti-inflammatory and antioxidant molecules. Natural products are structurally optimized by evolution to serve particular biological functions, including the regulation of endogenous defense mechanisms and interaction with other organisms. This property explains their relevance for infectious diseases and cancer. Recently, among the various natural substances, polyphenols from extra virgin olive oil (EVOO), an important element of the Mediterranean diet, have aroused growing interest. Extensive studies have shown the potent therapeutic effects of these bioactive molecules against a series of chronic diseases, such as cardiovascular diseases, diabetes, neurodegenerative disorders and cancer. This review begins from the chemical structure, abundance and bioavailability of the main EVOO polyphenols to highlight the effects and the possible molecular mechanism(s) of action of these compounds against inflammation and oxidation, in vitro and in vivo. In addition, the mechanisms of inhibition of molecular signaling pathways activated by oxidative stress by EVOO polyphenols are discussed, together with their possible roles in inflammation-mediated chronic disorders, also taking into account meta-analysis of population studies and clinical trials.
Collapse
Affiliation(s)
- Monica Bucciantini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence 50134, Italy; (M.L.); (M.S.)
- Correspondence:
| | - Manuela Leri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence 50134, Italy; (M.L.); (M.S.)
| | - Pamela Nardiello
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence 50134, Italy; (P.N.); (F.C.)
| | - Fiorella Casamenti
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence 50134, Italy; (P.N.); (F.C.)
| | - Massimo Stefani
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence 50134, Italy; (M.L.); (M.S.)
| |
Collapse
|
17
|
Westfall S, Caracci F, Estill M, Frolinger T, Shen L, Pasinetti GM. Chronic Stress-Induced Depression and Anxiety Priming Modulated by Gut-Brain-Axis Immunity. Front Immunol 2021; 12:670500. [PMID: 34248950 PMCID: PMC8264434 DOI: 10.3389/fimmu.2021.670500] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 06/08/2021] [Indexed: 12/18/2022] Open
Abstract
Chronic stress manifests as depressive- and anxiety-like behavior while recurrent stress elicits disproportionate behavioral impairments linked to stress-induced immunological priming. The gut-brain-microbiota-axis is a promising therapeutic target for stress-induced behavioral impairments as it simultaneously modulates peripheral and brain immunological landscapes. In this study, a combination of probiotics and prebiotics, known as a synbiotic, promoted behavioral resilience to chronic and recurrent stress by normalizing gut microbiota populations and promoting regulatory T cell (Treg) expansion through modulation of ileal innate lymphoid cell (ILC)3 activity, an impact reflecting behavioral responses better than limbic brain region neuroinflammation. Supporting this conclusion, a multivariate machine learning model correlatively predicted a cross-tissue immunological signature of stress-induced behavioral impairment where the ileal Treg/T helper17 cell ratio associated to hippocampal chemotactic chemokine and prefrontal cortex IL-1β production in the context of stress-induced behavioral deficits. In conclusion, stress-induced behavioral impairments depend on the gut-brain-microbiota-axis and through ileal immune regulation, synbiotics attenuate the associated depressive- and anxiety-like behavior.
Collapse
Affiliation(s)
- Susan Westfall
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Francesca Caracci
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Molly Estill
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Tal Frolinger
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Li Shen
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Giulio M. Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
| |
Collapse
|
18
|
Lamas B, Hernandez-Galan L, Galipeau HJ, Constante M, Clarizio A, Jury J, Breyner NM, Caminero A, Rueda G, Hayes CL, McCarville JL, Bermudez Brito M, Planchais J, Rolhion N, Murray JA, Langella P, Loonen LMP, Wells JM, Bercik P, Sokol H, Verdu EF. Aryl hydrocarbon receptor ligand production by the gut microbiota is decreased in celiac disease leading to intestinal inflammation. Sci Transl Med 2021; 12:12/566/eaba0624. [PMID: 33087499 DOI: 10.1126/scitranslmed.aba0624] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 02/24/2020] [Accepted: 06/02/2020] [Indexed: 12/15/2022]
Abstract
Metabolism of tryptophan by the gut microbiota into derivatives that activate the aryl hydrocarbon receptor (AhR) contributes to intestinal homeostasis. Many chronic inflammatory conditions, including celiac disease involving a loss of tolerance to dietary gluten, are influenced by cues from the gut microbiota. We investigated whether AhR ligand production by the gut microbiota could influence gluten immunopathology in nonobese diabetic (NOD) mice expressing DQ8, a celiac disease susceptibility gene. NOD/DQ8 mice, exposed or not exposed to gluten, were subjected to three interventions directed at enhancing AhR pathway activation. These included a high-tryptophan diet, gavage with Lactobacillus reuteri that produces AhR ligands or treatment with an AhR agonist. We investigated intestinal permeability, gut microbiota composition determined by 16S rRNA gene sequencing, AhR pathway activation in intestinal contents, and small intestinal pathology and inflammatory markers. In NOD/DQ8 mice, a high-tryptophan diet modulated gut microbiota composition and enhanced AhR ligand production. AhR pathway activation by an enriched tryptophan diet, treatment with the AhR ligand producer L. reuteri, or pharmacological stimulation using 6-formylindolo (3,2-b) carbazole (Ficz) decreased immunopathology in NOD/DQ8 mice exposed to gluten. We then determined AhR ligand production by the fecal microbiota and AhR activation in patients with active celiac disease compared to nonceliac control individuals. Patients with active celiac disease demonstrated reduced AhR ligand production and lower intestinal AhR pathway activation. These results highlight gut microbiota-dependent modulation of the AhR pathway in celiac disease and suggest a new therapeutic strategy for treating this disorder.
Collapse
Affiliation(s)
- Bruno Lamas
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Leticia Hernandez-Galan
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Heather J Galipeau
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Marco Constante
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Alexandra Clarizio
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jennifer Jury
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Natalia M Breyner
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Alberto Caminero
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Gaston Rueda
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Christina L Hayes
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Justin L McCarville
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Miriam Bermudez Brito
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Julien Planchais
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Nathalie Rolhion
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Service de Gastroenterologie, F-75012 Paris, France
| | - Joseph A Murray
- Division of Gastroenterology and Hepatology, Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Philippe Langella
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Linda M P Loonen
- Host-Microbe Interactomics, Animal Sciences Group, Wageningen University, Wageningen, Netherlands
| | - Jerry M Wells
- Host-Microbe Interactomics, Animal Sciences Group, Wageningen University, Wageningen, Netherlands
| | - Premysl Bercik
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Harry Sokol
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France. .,Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Service de Gastroenterologie, F-75012 Paris, France
| | - Elena F Verdu
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
19
|
Huang Z, Schoones T, Wells JM, Fogliano V, Capuano E. Substrate-Driven Differences in Tryptophan Catabolism by Gut Microbiota and Aryl Hydrocarbon Receptor Activation. Mol Nutr Food Res 2021; 65:e2100092. [PMID: 33964185 PMCID: PMC8365636 DOI: 10.1002/mnfr.202100092] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/01/2021] [Indexed: 12/17/2022]
Abstract
SCOPE This study aims to investigate the effect of tryptophan sources on tryptophan catabolism by gut microbiota and the aryl hydrocarbon receptor (AhR) activation. METHODS AND RESULTS Four substrates (free tryptophan, soybean protein, single and clustered soybean cells) containing an equimolar amount of tryptophan, but with a different bioaccessibility are studied using in vitro batch fermentation. Tryptophan catabolites are identified by LC-MS/MS. AhR activity is measured by HepG2-Lucia AhR reporter cells. The total amount of tryptophan-derived catabolites increases with decreasing level of substrate complexity. Indole is the major catabolite produced from tryptophan and it is the most abundant in the free tryptophan fermentation. Indole-3-acetic acid and indole-3-aldehyde are abundantly generated in the soybean protein fermentation. The soybean cell fermentation produced high concentrations of tryptamine. Interestingly, large amounts of short-chain fatty acids (SCFAs) are also found in the soybean cell and protein fermentation. Both tryptophan-derived catabolites and SCFAs are able to increase AhR reporter activity over time in all four groups. CONCLUSION This study illustrates that bacterial catabolism of tryptophan and resulting AhR activation in the gut is modulated by the food matrix, suggesting a role for food design to improve gut health.
Collapse
Affiliation(s)
- Zhan Huang
- Food Quality and Design Group, Department of Agrotechnology and Food Sciences, Wageningen University, P.O. Box 17, Wageningen, 6700 AA, The Netherlands.,Host-Microbe Interactomics Group, Department of Animal Sciences, Wageningen University, P.O. Box 17, Wageningen, 6700 AA, The Netherlands
| | - Tessa Schoones
- Food Quality and Design Group, Department of Agrotechnology and Food Sciences, Wageningen University, P.O. Box 17, Wageningen, 6700 AA, The Netherlands
| | - Jerry M Wells
- Host-Microbe Interactomics Group, Department of Animal Sciences, Wageningen University, P.O. Box 17, Wageningen, 6700 AA, The Netherlands
| | - Vincenzo Fogliano
- Food Quality and Design Group, Department of Agrotechnology and Food Sciences, Wageningen University, P.O. Box 17, Wageningen, 6700 AA, The Netherlands
| | - Edoardo Capuano
- Food Quality and Design Group, Department of Agrotechnology and Food Sciences, Wageningen University, P.O. Box 17, Wageningen, 6700 AA, The Netherlands
| |
Collapse
|
20
|
Millman JF, Okamoto S, Teruya T, Uema T, Ikematsu S, Shimabukuro M, Masuzaki H. Extra-virgin olive oil and the gut-brain axis: influence on gut microbiota, mucosal immunity, and cardiometabolic and cognitive health. Nutr Rev 2021; 79:1362-1374. [PMID: 33576418 PMCID: PMC8581649 DOI: 10.1093/nutrit/nuaa148] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Extra-virgin olive oil (EVOO), a popular functional food and major source of fat in the Mediterranean diet, possesses a variety of healthful components, including monounsaturated fatty acids and bioactive phenolic compounds that, individually and collectively, exert beneficial effects on cardiometabolic markers of health and act as neuroprotective agents through their anti-inflammatory and antioxidant activities. The gut microbiota and health of the intestinal environment are now considered important factors in the development of obesity, metabolic disease, and even certain neurodegenerative conditions via the gut-brain axis. Recently, data are emerging which demonstrate that the health-promoting benefits of EVOO may also extend to the gut microbiota. In this review, we aimed to examine findings from recent studies regarding the impact of EVOO on gut microbiota and intestinal health and explore how modulations in composition of gut microbiota, production of microbially produced products, and activity and functioning of the mucosal immune system may lead to favorable outcomes in cardiovascular, metabolic, and cognitive health.
Collapse
Affiliation(s)
- Jasmine F Millman
- J.F. Millman, S. Okamoto, T. Taiki, T. Uema, and H. Masuzaki are with the Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. S. Ikematsu is with the Department of Bioresources Engineering, National Institute of Technology, Okinawa College, Okinawa, Japan. M. Shimabukuro is with the Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University, Fukushima, Japan
| | - Shiki Okamoto
- J.F. Millman, S. Okamoto, T. Taiki, T. Uema, and H. Masuzaki are with the Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. S. Ikematsu is with the Department of Bioresources Engineering, National Institute of Technology, Okinawa College, Okinawa, Japan. M. Shimabukuro is with the Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University, Fukushima, Japan
| | - Taiki Teruya
- J.F. Millman, S. Okamoto, T. Taiki, T. Uema, and H. Masuzaki are with the Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. S. Ikematsu is with the Department of Bioresources Engineering, National Institute of Technology, Okinawa College, Okinawa, Japan. M. Shimabukuro is with the Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University, Fukushima, Japan
| | - Tsugumi Uema
- J.F. Millman, S. Okamoto, T. Taiki, T. Uema, and H. Masuzaki are with the Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. S. Ikematsu is with the Department of Bioresources Engineering, National Institute of Technology, Okinawa College, Okinawa, Japan. M. Shimabukuro is with the Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University, Fukushima, Japan
| | - Shinya Ikematsu
- J.F. Millman, S. Okamoto, T. Taiki, T. Uema, and H. Masuzaki are with the Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. S. Ikematsu is with the Department of Bioresources Engineering, National Institute of Technology, Okinawa College, Okinawa, Japan. M. Shimabukuro is with the Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University, Fukushima, Japan
| | - Michio Shimabukuro
- J.F. Millman, S. Okamoto, T. Taiki, T. Uema, and H. Masuzaki are with the Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. S. Ikematsu is with the Department of Bioresources Engineering, National Institute of Technology, Okinawa College, Okinawa, Japan. M. Shimabukuro is with the Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University, Fukushima, Japan
| | - Hiroaki Masuzaki
- J.F. Millman, S. Okamoto, T. Taiki, T. Uema, and H. Masuzaki are with the Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. S. Ikematsu is with the Department of Bioresources Engineering, National Institute of Technology, Okinawa College, Okinawa, Japan. M. Shimabukuro is with the Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
21
|
Zhu YD, Guan XQ, Chen J, Peng S, Finel M, Zhao YY, Wang RM, Bi HC, Lei M, Wang DD, Ge GB. Neobavaisoflavone Induces Bilirubin Metabolizing Enzyme UGT1A1 via PPARα and PPARγ. Front Pharmacol 2021; 11:628314. [PMID: 33628187 PMCID: PMC7897654 DOI: 10.3389/fphar.2020.628314] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/24/2020] [Indexed: 12/30/2022] Open
Abstract
UDP-glucuronosyltransferase 1A1 (UGT1A1) is an essential enzyme in mammals that is responsible for detoxification and metabolic clearance of the endogenous toxin bilirubin and a variety of xenobiotics, including some crucial therapeutic drugs. Discovery of potent and safe UGT1A1 inducers will provide an alternative therapy for ameliorating hyperbilirubinaemia and drug-induced hepatoxicity. This study aims to find efficacious UGT1A1 inducer(s) from natural flavonoids, and to reveal the mechanism involved in up-regulating of this key conjugative enzyme by the flavonoid(s) with strong UGT1A1 induction activity. Among all the tested flavonoids, neobavaisoflavone (NBIF) displayed the most potent UGT1A1 induction activity, while its inductive effects were confirmed by both western blot and glucuronidation activity assays. A panel of nuclear receptor reporter assays demonstrated that NBIF activated PPARα and PPARγ in a dose-dependent manner. Meanwhile, we also found that NBIF could up-regulate the expression of PPARα and PPARγ in hepatic cells, suggesting that the induction of UGT1A1 by NBIF was mainly mediated by PPARs. In silico simulations showed that NBIF could stably bind on pocket II of PPARα and PPARγ. Collectively, our results demonstrated that NBIF is a natural inducer of UGT1A1, while this agent induced UGT1A1 mainly via activating and up-regulating PPARα and PPARγ. These findings suggested that NBIF can be used as a promising lead compound for the development of more efficacious UGT1A1 inducers to treat hyperbilirubinaemia and UGT1A1-associated drug toxicities.
Collapse
Affiliation(s)
- Ya-Di Zhu
- Trauma Emergency Center, The Seventh Affiliated People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Qing Guan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sheng Peng
- Trauma Emergency Center, The Seventh Affiliated People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Moshe Finel
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Ying-Yuan Zhao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Rui-Min Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Hui-Chang Bi
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ming Lei
- Trauma Emergency Center, The Seventh Affiliated People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan-Dan Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang-Bo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
22
|
Rovalino-Córdova AM, Fogliano V, Capuano E. In vitro colonic fermentation of red kidney beans depends on cotyledon cells integrity and microbiota adaptation. Food Funct 2021; 12:4983-4994. [PMID: 34100499 DOI: 10.1039/d1fo00321f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the present study we investigated the effect of cellular integrity on microbial utilization of proteins and carbohydrates by gut microbiota. Cotyledon cells from red kidney beans with different levels of structural integrity were fermented in-vitro by microbial communities previously adapted to the conditions of ascending, transverse and descending colon. The effect of bacterial adaptation to substrate was also assessed by using microbiota exposed to a diet rich in bean cells. Microscopy analyses indicate that cell integrity was maintained during fermentation. The amount of gas generated and the rate of total gas production was higher in broken cells compared to intact cells which suggest a faster and more extensive utilization of nutrients when cell wall is broken. A significantly higher butyric and propionic acid level was detected in broken cells at the end of the fermentation. Moreover, adapted bacterial communities were more efficient in fermenting bean cells where higher amounts of butyrate were produced in all colon regions independently of sample integrity. Bacterial communities of the distal colon appeared to be the most efficient in carbohydrate and protein fermentation as witnessed by the higher levels of gas, and short chain fatty acids. It was also found that cell integrity and adaptation to bean cells modulate the hierarchy of nutrient utilization, with non-starch polysaccharides preferred over starch and proteins by microbiota exposed to bean cells. Our results demonstrated that structural aspects of foods, such as cell integrity in plant tissues, may modulate nutrients utilization by gut microbiota.
Collapse
Affiliation(s)
- Ana M Rovalino-Córdova
- Food Quality and Design, Wageningen University, P.O. Box 17, 6700 AA Wageningen, The Netherlands.
| | - Vincenzo Fogliano
- Food Quality and Design, Wageningen University, P.O. Box 17, 6700 AA Wageningen, The Netherlands.
| | - Edoardo Capuano
- Food Quality and Design, Wageningen University, P.O. Box 17, 6700 AA Wageningen, The Netherlands.
| |
Collapse
|
23
|
Diotallevi C, Fava F, Gobbetti M, Tuohy K. Healthy dietary patterns to reduce obesity-related metabolic disease: polyphenol-microbiome interactions unifying health effects across geography. Curr Opin Clin Nutr Metab Care 2020; 23:437-444. [PMID: 32941185 DOI: 10.1097/mco.0000000000000697] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The spread of the Western lifestyle across the globe has led to a pandemic in obesity-related metabolic disease. The Mediterranean diet (MedDiet), Okinawa diet (OkD) and Nordic diet, derived from very different regions of the world and culinary traditions, have a large whole plant food component and are associated with reduced disease risk. This review focuses on polyphenol : microbiome interactions as one possible common mechanistic driver linking the protective effects whole plant foods against metabolic disease across healthy dietary patterns irrespective of geography. RECENT FINDINGS Although mechanistic evidence in humans is still scarce, animal studies suggest that polyphenol or polyphenol rich foods induce changes within the gut microbiota and its metabolic output of trimethylamine N-oxide, short-chain fatty acids, bile acids and small phenolic acids. These cross-kingdom signaling molecules regulate mammalian lipid and glucose homeostasis, inflammation and energy storage or thermogenesis, physiological processes determining obesity-related metabolic and cardiovascular disease risk. However, it appears that where in the intestine metabolites are produced, the microbiota communities involved, and interactions between the metabolites themselves, can all influence physiological responses, highlighting the need for a greater understanding of the kinetics and site of production of microbial metabolites within the gut. SUMMARY Interactions between polyphenols and metabolites produced by the gut microbiota are emerging as a possible unifying protective mechanism underpinning diverse healthy dietary patterns signaling across culinary traditions, across geography and across domains of life.
Collapse
Affiliation(s)
- Camilla Diotallevi
- Faculty of Science and Technology, Freie Universität Bozen-Libera Università di Bolzano, Bolzano
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, Trento, Italy
| | - Francesca Fava
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, Trento, Italy
| | - Marco Gobbetti
- Faculty of Science and Technology, Freie Universität Bozen-Libera Università di Bolzano, Bolzano
| | - Kieran Tuohy
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, Trento, Italy
| |
Collapse
|
24
|
Effect of bean structure on microbiota utilization of plant nutrients: An in-vitro study using the simulator of the human intestinal microbial ecosystem (SHIME®). J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104087] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
25
|
Ninfali P, Antonelli A, Magnani M, Scarpa ES. Antiviral Properties of Flavonoids and Delivery Strategies. Nutrients 2020; 12:nu12092534. [PMID: 32825564 PMCID: PMC7551920 DOI: 10.3390/nu12092534] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/14/2020] [Accepted: 08/18/2020] [Indexed: 12/18/2022] Open
Abstract
This review summarizes the latest advancements in phytochemicals as functional antiviral agents. We focused on flavonoids, like apigenin, vitexin, quercetin, rutin and naringenin, which have shown a wide range of biological effects including antiviral activities. The molecular mechanisms of their antiviral effects mainly consist in the inhibition of viral neuraminidase, proteases and DNA/RNA polymerases, as well as in the modification of various viral proteins. Mixtures of different flavonoids or combination of flavonoids with antiviral synthetic drugs provide an enhancement of their antiviral effects. Recent strategies in drug delivery significantly contribute to overcoming the low bioavailability of flavonoids. Frequent viral infections worldwide have led to the need for new effective antiviral agents, which can be identified among the various phytochemicals. In this light, screening the antiviral activities of a cocktail of flavonoids would be advantageous in order to prevent viral infections and improve current antiviral therapies.
Collapse
Affiliation(s)
| | | | - Mauro Magnani
- Correspondence: (M.M.); (E.S.S.); Tel.: +39-0722-305-211 (M.M.); +39-0722-305-252 (E.S.S.)
| | | |
Collapse
|
26
|
Tsvetikova SA, Koshel EI. Microbiota and cancer: host cellular mechanisms activated by gut microbial metabolites. Int J Med Microbiol 2020; 310:151425. [PMID: 32423739 DOI: 10.1016/j.ijmm.2020.151425] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 03/25/2020] [Accepted: 04/13/2020] [Indexed: 12/13/2022] Open
|
27
|
Koper JEB, Kortekaas M, Loonen LMP, Huang Z, Wells JM, Gill CIR, Pourshahidi LK, McDougall G, Rowland I, Pereira-Caro G, Fogliano V, Capuano E. Aryl hydrocarbon Receptor activation during in vitro and in vivo digestion of raw and cooked broccoli (brassica oleracea var. Italica). Food Funct 2020; 11:4026-4037. [PMID: 32323699 DOI: 10.1039/d0fo00472c] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Broccoli is rich in glucosinolates, which can be converted upon chewing and processing into Aryl hydrocarbon Receptor (AhR) ligands. Activation of AhR plays an important role in overall gut homeostasis but the role of broccoli processing on the generation of AhR ligands is still largely unknown. In this study, the effects of temperature, cooking method (steaming versus boiling), gastric pH and further digestion of broccoli on AhR activation were investigated in vitro and in ileostomy subjects. For the in vitro study, raw, steamed (t = 3 min and t = 6 min) and boiled (t = 3 min and t = 6 min) broccoli were digested in vitro with different gastric pH. In the in vivo ileostomy study, 8 subjects received a broccoli soup or a broccoli soup plus an exogenous myrosinase source. AhR activation was measured in both in vitro and in vivo samples by using HepG2-Lucia™ AhR reporter cells. Cooking broccoli reduced the AhR activation measured after gastric digestion in vitro, but no effect of gastric pH was found. Indole AhR ligands were not detected or detected at very low levels both after intestinal in vitro digestion and in the ileostomy patient samples, which resulted in no AhR activation. This suggests that the evaluation of the relevance of glucosinolates for AhR modulation in the gut cannot prescind from the way broccoli is processed, and that broccoli consumption does not necessarily produce substantial amounts of AhR ligands in the large intestine.
Collapse
Affiliation(s)
- Jonna E B Koper
- Wageningen University, Department of Agrotechnology & Food Sciences, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Enhanced production of Th1- and Th2-type antibodies and induction of regulatory T cells in mice by oral administration of Cyclopia extracts with similar phenolic composition to honeybush herbal tea. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103704] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
29
|
Fois CAM, Le TYL, Schindeler A, Naficy S, McClure DD, Read MN, Valtchev P, Khademhosseini A, Dehghani F. Models of the Gut for Analyzing the Impact of Food and Drugs. Adv Healthc Mater 2019; 8:e1900968. [PMID: 31592579 DOI: 10.1002/adhm.201900968] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/30/2019] [Indexed: 12/16/2022]
Abstract
Models of the human gastrointestinal tract (GIT) can be powerful tools for examining the biological interactions of food products and pharmaceuticals. This can be done under normal healthy conditions or using models of disease-many of which have no curative therapy. This report outlines the field of gastrointestinal modeling, with a particular focus on the intestine. Traditional in vivo animal models are compared to a range of in vitro models. In vitro systems are elaborated over time, recently culminating with microfluidic intestines-on-chips (IsOC) and 3D bioengineered models. Macroscale models are also reviewed for their important contribution in the microbiota studies. Lastly, it is discussed how in silico approaches may have utility in predicting and interpreting experimental data. The various advantages and limitations of the different systems are contrasted. It is posited that only through complementary use of these models will salient research questions be able to be addressed.
Collapse
Affiliation(s)
- Chiara Anna Maria Fois
- School of Chemical and Biomolecular Engineering Centre for Advanced Food Enginomics University of Sydney Sydney NSW 2006 Australia
| | - Thi Yen Loan Le
- School of Chemical and Biomolecular Engineering Centre for Advanced Food Enginomics University of Sydney Sydney NSW 2006 Australia
| | - Aaron Schindeler
- School of Chemical and Biomolecular Engineering Centre for Advanced Food Enginomics University of Sydney Sydney NSW 2006 Australia
| | - Sina Naficy
- School of Chemical and Biomolecular Engineering Centre for Advanced Food Enginomics University of Sydney Sydney NSW 2006 Australia
| | - Dale David McClure
- School of Chemical and Biomolecular Engineering Centre for Advanced Food Enginomics University of Sydney Sydney NSW 2006 Australia
| | - Mark Norman Read
- School of Chemical and Biomolecular Engineering Centre for Advanced Food Enginomics University of Sydney Sydney NSW 2006 Australia
| | - Peter Valtchev
- School of Chemical and Biomolecular Engineering Centre for Advanced Food Enginomics University of Sydney Sydney NSW 2006 Australia
| | - Ali Khademhosseini
- Department of Chemical and Biomolecular Engineering Department of Bioengineering Department of Radiology California NanoSystems Institute (CNSI) University of California Los Angeles CA 90095 USA
| | - Fariba Dehghani
- School of Chemical and Biomolecular Engineering Centre for Advanced Food Enginomics University of Sydney Sydney NSW 2006 Australia
| |
Collapse
|
30
|
The Dynamic Interplay between the Gut Microbiota and Autoimmune Diseases. J Immunol Res 2019; 2019:7546047. [PMID: 31772949 PMCID: PMC6854958 DOI: 10.1155/2019/7546047] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/14/2019] [Indexed: 12/11/2022] Open
Abstract
The human gut-resident commensal microbiota is a unique ecosystem associated with various bodily functions, especially immunity. Gut microbiota dysbiosis plays a crucial role in autoimmune disease pathogenesis as well as in bowel-related diseases. However, the role of the gut microbiota, which causes or influences systemic immunity in autoimmune diseases, remains elusive. Aryl hydrocarbon receptor, a ligand-activated transcription factor, is a master moderator of host-microbiota interactions because it shapes the immune system and impacts host metabolism. In addition, treatment optimization while minimizing potential adverse effects in autoimmune diseases remains essential, and modulation of the gut microbiota constitutes a potential clinical therapy. Here, we present evidence linking gut microbiota dysbiosis with autoimmune mechanisms involved in disease development to identify future effective approaches based on the gut microbiota for preventing autoimmune diseases.
Collapse
|
31
|
Abstract
The human gut-resident commensal microbiota is a unique ecosystem associated with various bodily functions, especially immunity. Gut microbiota dysbiosis plays a crucial role in autoimmune disease pathogenesis as well as in bowel-related diseases. However, the role of the gut microbiota, which causes or influences systemic immunity in autoimmune diseases, remains elusive. Aryl hydrocarbon receptor, a ligand-activated transcription factor, is a master moderator of host-microbiota interactions because it shapes the immune system and impacts host metabolism. In addition, treatment optimization while minimizing potential adverse effects in autoimmune diseases remains essential, and modulation of the gut microbiota constitutes a potential clinical therapy. Here, we present evidence linking gut microbiota dysbiosis with autoimmune mechanisms involved in disease development to identify future effective approaches based on the gut microbiota for preventing autoimmune diseases.
Collapse
|
32
|
Gheorghe CE, Martin JA, Manriquez FV, Dinan TG, Cryan JF, Clarke G. Focus on the essentials: tryptophan metabolism and the microbiome-gut-brain axis. Curr Opin Pharmacol 2019; 48:137-145. [PMID: 31610413 DOI: 10.1016/j.coph.2019.08.004] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 08/26/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
Abstract
The gut-brain axis is a bidirectional communication system between the central nervous system and the gastrointestinal tract, in which serotonin (5-HT) functions as a key neurotransmitter. Recent research has increasingly concentrated on tryptophan, the precursor to 5-HT and on the microbial regulation of tryptophan metabolism, with an emphasis on host-microbe control over kynurenine pathway metabolism and microbial-specific pathways that generate bioactive tryptophan metabolites. Here, we critically assess recent progress made towards a mechanistic understanding of the microbial regulation of tryptophan metabolism and microbiota-gut-brain axis homeostasis highlighting the role tryptophan metabolism plays in preclinical and clinical neuroscience and in the challenge to improve our understanding of how perturbed tryptophan metabolism contributes to stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Cassandra Elise Gheorghe
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jason A Martin
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Francisca Villalobos Manriquez
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland; INFANT Research Centre, University College Cork, Cork, Ireland.
| |
Collapse
|
33
|
Yang T, Feng YL, Chen L, Vaziri ND, Zhao YY. Dietary natural flavonoids treating cancer by targeting aryl hydrocarbon receptor. Crit Rev Toxicol 2019; 49:445-460. [PMID: 31433724 DOI: 10.1080/10408444.2019.1635987] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The role of aryl hydrocarbon receptor (AhR) as a ligand-activated transcription factor in the field of cancer has gradually been unveiled. A strong body of evidence indicated that AhR is implicated in cell proliferation and apoptosis, immune metabolism and other processes, which further affected tumor growth, survival, migration, and invasion. Therefore, AhR targeted therapy may become a new method for cancer treatment and provide a new direction for clinical tumor treatment. Astonishingly, the largest source of exposure of animals and humans to AhR ligands (synthetic and natural) comes from the diet. Myriad studies have described that various natural dietary chemicals can directly activate and/or inhibit the AhR signaling pathway. Of note, numerous natural products contribute to AhR active, of which dietary flavonoids are the largest class of natural AhR ligands. As interest in AhR and its ligands increases, it seems sensible to summarize current research on these ligands. In this review, we highlight the role of AhR in tumorigenesis and focus on the double effect of AhR in cancer therapy. We explored the molecular mechanism of AhR ligands on cancer through a few AhR agonists/antagonists currently in clinical practice. Ultimately, we summarize and highlight the latest progression of dietary flavonoids as AhR ligands in cancer inhibition, including the limitations and deficiencies of it in clinical research. This review will offer a comprehensive understanding of AhR and its dietary ligands which may dramatically pave the way for targeted cancer treatment.
Collapse
Affiliation(s)
- Tian Yang
- Faculty of Life Science & Medicine, Northwest University, Xi'an, China
| | - Ya-Long Feng
- Faculty of Life Science & Medicine, Northwest University, Xi'an, China
| | - Lin Chen
- Faculty of Life Science & Medicine, Northwest University, Xi'an, China
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Ying-Yong Zhao
- Faculty of Life Science & Medicine, Northwest University, Xi'an, China
| |
Collapse
|
34
|
Papillo VA, Arlorio M, Locatelli M, Fuso L, Pellegrini N, Fogliano V. In vitro evaluation of gastro-intestinal digestion and colonic biotransformation of curcuminoids considering different formulations and food matrices. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.05.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
35
|
Biasutto L, Mattarei A, La Spina M, Azzolini M, Parrasia S, Szabò I, Zoratti M. Strategies to target bioactive molecules to subcellular compartments. Focus on natural compounds. Eur J Med Chem 2019; 181:111557. [PMID: 31374419 DOI: 10.1016/j.ejmech.2019.07.060] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/04/2019] [Accepted: 07/21/2019] [Indexed: 02/06/2023]
Abstract
Many potential pharmacological targets are present in multiple subcellular compartments and have different pathophysiological roles depending on location. In these cases, selective targeting of a drug to the relevant subcellular domain(s) may help to sharpen its impact by providing topological specificity, thus limiting side effects, and to concentrate the compound where needed, thus increasing its effectiveness. We review here the state of the art in precision subcellular delivery. The major approaches confer "homing" properties to the active principle via permanent or reversible (in pro-drug fashion) modifications, or through the use of special-design nanoparticles or liposomes to ferry a drug(s) cargo to its desired destination. An assortment of peptides, substituents with delocalized positive charges, custom-blended lipid mixtures, pH- or enzyme-sensitive groups provide the main tools of the trade. Mitochondria, lysosomes and the cell membrane may be mentioned as the fronts on which the most significant advances have been made. Most of the examples presented here have to do with targeting natural compounds - in particular polyphenols, known as pleiotropic agents - to one or the other subcellular compartment.
Collapse
Affiliation(s)
- Lucia Biasutto
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121, Padova, Italy; Dept. Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121, Padova, Italy.
| | - Andrea Mattarei
- Dept. Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131, Padova, Italy
| | - Martina La Spina
- Dept. Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121, Padova, Italy
| | - Michele Azzolini
- Dept. Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121, Padova, Italy
| | - Sofia Parrasia
- Dept. Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121, Padova, Italy
| | - Ildikò Szabò
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121, Padova, Italy; Dept. Biology, University of Padova, Viale G. Colombo 3, 35121, Padova, Italy
| | - Mario Zoratti
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121, Padova, Italy; Dept. Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121, Padova, Italy
| |
Collapse
|
36
|
de Souza JV, Reznikov S, Zhu R, Bronowska AK. Druggability assessment of mammalian Per-Arnt-Sim [PAS] domains using computational approaches. MEDCHEMCOMM 2019; 10:1126-1137. [PMID: 31391885 PMCID: PMC6640724 DOI: 10.1039/c9md00148d] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 05/01/2019] [Indexed: 01/10/2023]
Abstract
Per-Arnt-Sim (PAS) domains are key regions that occur in different regulatory proteins from all kingdoms of life. PAS domains show a remarkably conserved structural scaffold, despite a highly variable primary sequence. In this study we have attempted to address some of the gaps in knowledge regarding the druggability of PAS-A domains, differences in structure and dynamics within the PAS domain family and how this affects the druggability potential, as well as give insight into the druggability of steroid receptor coactivators and putative binding modes of the NCOA1. Investigations were performed through a range of computational methods including molecular docking studies, atomistic molecular dynamics simulations, and hotspot mapping. Atomistic molecular dynamics simulations show that the function of the AhR PAS-B domain is regulated by the dynamics of the highly conserved tyrosine Y322 residue, which acts as a "gatekeeper" controlling the access to the binding cavity and finely tuning the binding affinity. Furthermore, the transition between the partially unfolded and helical conformation of the loop1 segment within PAS-B domains was shown to be essential for the generation of "druggable" sites, especially for the NCOA1 PAS-B domain. Finally, our simulations indicated the undruggability of PAS-A domains, caused by the inherent characteristics of their putative binding sites. In conclusion, this work emphasises the role of intrinsic dynamics in tuning the druggability of PAS-B domains and shows that PAS-B domains of steroid receptor coactivators, such as NCOA1, can be targeted by small molecule ligands, which highlights the potential of developing new therapeutics designed to target these coactivators using structure-based approaches.
Collapse
Affiliation(s)
- João V de Souza
- School of Natural and Environmental Sciences , Newcastle University , NE1 7RU Newcastle , UK .
| | - Sylvia Reznikov
- School of Natural and Environmental Sciences , Newcastle University , NE1 7RU Newcastle , UK .
| | - Ruidi Zhu
- School of Natural and Environmental Sciences , Newcastle University , NE1 7RU Newcastle , UK .
| | - Agnieszka K Bronowska
- School of Natural and Environmental Sciences , Newcastle University , NE1 7RU Newcastle , UK .
| |
Collapse
|
37
|
Koper JEB, Loonen LMP, Wells JM, Troise AD, Capuano E, Fogliano V. Polyphenols and Tryptophan Metabolites Activate the Aryl Hydrocarbon Receptor in an in vitro Model of Colonic Fermentation. Mol Nutr Food Res 2018; 63:e1800722. [PMID: 30443985 PMCID: PMC6588005 DOI: 10.1002/mnfr.201800722] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/07/2018] [Indexed: 12/23/2022]
Abstract
Scope Many dietary phytochemicals have been reported to promote gut health. Specific dietary phytochemicals, such as luteolin, as well as specific microbial metabolites of tryptophan are ligands of the aryl hydrocarbon receptor (AhR), which plays a role in immunity and homeostasis of the gut barrier. Here, the fate of luteolin during colonic fermentation and the contribution of tryptophan metabolites to AhR activity in different parts of the colon are investigated. Methods and results Several polyphenols are screened for AhR activation and oregano, containing the ligand luteolin, is added to batch cultures of human microbiota from the distal colon. Luteolin is rapidly metabolized, with no measurable increase in AhR activity. In the second experiment, using the Simulator of the Human Intestinal Microbial Ecosystem (SHIME), not all luteolin is metabolized in the ascending colon, but disappear rapidly in the transverse colon. The greatest AhR activity is due to microbiota‐derived metabolites of tryptophan, particularly in the descending colon. Conclusions Luteolin in food is rapidly metabolized in the transverse colon. Tryptophan metabolism by the microbiota in the colon contributes substantially to the pool of lumen metabolites that can activate the AhR.
Collapse
Affiliation(s)
- Jonna E B Koper
- Department of Agrotechnology & Food Sciences, Wageningen University, Wageningen, 6708 WG, The Netherlands.,Department of Animal Sciences, Wageningen University, Wageningen, 6708 WG, The Netherlands
| | - Linda M P Loonen
- Department of Animal Sciences, Wageningen University, Wageningen, 6708 WG, The Netherlands
| | - Jerry M Wells
- Department of Animal Sciences, Wageningen University, Wageningen, 6708 WG, The Netherlands
| | - Antonio Dario Troise
- Department of Agricultural Sciences, University of Naples "Federico II", Portici, 80055, Italy
| | - Edoardo Capuano
- Department of Agrotechnology & Food Sciences, Wageningen University, Wageningen, 6708 WG, The Netherlands
| | - Vincenzo Fogliano
- Department of Agrotechnology & Food Sciences, Wageningen University, Wageningen, 6708 WG, The Netherlands
| |
Collapse
|