1
|
Seong H, Han SH, Kim G, Han NS. Viability and probiotic activity of Lactiplantibacillus plantarum PMO08 in human gastrointestinal tract analyzed by in vitro gut model. Food Sci Biotechnol 2024; 33:2223-2231. [PMID: 39130653 PMCID: PMC11315860 DOI: 10.1007/s10068-024-01622-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 08/13/2024] Open
Abstract
This study aimed to evaluate the survivability of Lactiplantibacillus plantarum PMO08 in the human gastrointestinal tract and its adaptability in the colon using in vitro models. After exposure to gastric and small intestinal conditions, the majority (92.70 ± 1.14%) of PMO08 was found to be damaged, as determined by confocal microscopy and flow cytometry. During in vitro colonic fermentation, PMO08 not only increased abundance up to 0.47 ± 0.04% compared with the control sample (0.00 ± 0.00%) at 24 h but also facilitated the growth of beneficial or commensal bacteria, thereby increasing the α-diversity indices. Additionally, PMO08 significantly elevated the levels of short-chain fatty acids (SCFAs) and various organic acids. Our results demonstrate that PMO08 possesses moderate viability under gastrointestinal conditions but exhibits superior probiotic activity in the colon.
Collapse
Affiliation(s)
- Hyunbin Seong
- Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Chungbuk Republic of Korea
| | - Seung Hee Han
- Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Chungbuk Republic of Korea
| | - Geonhee Kim
- Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Chungbuk Republic of Korea
| | - Nam Soo Han
- Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Chungbuk Republic of Korea
| |
Collapse
|
2
|
Bavaro AR, Di Biase M, Linsalata V, D’Antuono I, Di Stefano V, Lonigro SL, Garbetta A, Valerio F, Melilli MG, Cardinali A. Potential Prebiotic Effect of Inulin-Enriched Pasta after In Vitro Gastrointestinal Digestion and Simulated Gut Fermentation. Foods 2024; 13:1815. [PMID: 38928756 PMCID: PMC11202534 DOI: 10.3390/foods13121815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/26/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
In the current study, the prebiotic potential of an innovative functional pasta enriched with 12% (w/w) inulin was investigated. To this aim, pasta was subjected to in vitro gastrointestinal digestion followed by simulated gut fermentation compared to the control pasta (CTRL) not containing inulin. The incorporation of inulin positively (p < 0.05) affected some organoleptic traits and the cooking quality of the final product, giving an overall score significantly higher than CTRL. The resultant essential amino acid content was similar in both pasta samples while the total protein content was lower in inulin-enriched pasta for the polymer substitution to durum wheat flour. The prebiotic potential of chicory inulin was preliminarily tested in in vitro experiments using seven probiotic strains and among them Lacticaseibacillus paracasei IMPC2.1 was selected for the simulated gut fermentation studies. The positive prebiotic activity score registered with the probiotic strain suggested the suitability of the inulin-enriched pasta with respect to acting as a prebiotic source favoring the growth of the probiotic strain and short chain fatty acid (SCFA) production. The present study contributes to broadening knowledge on the prebiotic efficacy of inulin when incorporated into a complex food matrix.
Collapse
Affiliation(s)
- Anna Rita Bavaro
- Institute of Sciences of Food Productions (ISPA), National Research Council (CNR), 70126 Bari, Italy; (A.R.B.); (M.D.B.); (V.L.); (I.D.); (S.L.L.); (A.G.); (A.C.)
| | - Mariaelena Di Biase
- Institute of Sciences of Food Productions (ISPA), National Research Council (CNR), 70126 Bari, Italy; (A.R.B.); (M.D.B.); (V.L.); (I.D.); (S.L.L.); (A.G.); (A.C.)
| | - Vito Linsalata
- Institute of Sciences of Food Productions (ISPA), National Research Council (CNR), 70126 Bari, Italy; (A.R.B.); (M.D.B.); (V.L.); (I.D.); (S.L.L.); (A.G.); (A.C.)
| | - Isabella D’Antuono
- Institute of Sciences of Food Productions (ISPA), National Research Council (CNR), 70126 Bari, Italy; (A.R.B.); (M.D.B.); (V.L.); (I.D.); (S.L.L.); (A.G.); (A.C.)
| | - Vita Di Stefano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy;
| | - Stella Lisa Lonigro
- Institute of Sciences of Food Productions (ISPA), National Research Council (CNR), 70126 Bari, Italy; (A.R.B.); (M.D.B.); (V.L.); (I.D.); (S.L.L.); (A.G.); (A.C.)
| | - Antonella Garbetta
- Institute of Sciences of Food Productions (ISPA), National Research Council (CNR), 70126 Bari, Italy; (A.R.B.); (M.D.B.); (V.L.); (I.D.); (S.L.L.); (A.G.); (A.C.)
| | - Francesca Valerio
- Institute of Sciences of Food Productions (ISPA), National Research Council (CNR), 70126 Bari, Italy; (A.R.B.); (M.D.B.); (V.L.); (I.D.); (S.L.L.); (A.G.); (A.C.)
| | - Maria Grazia Melilli
- Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), 95126 Catania, Italy
| | - Angela Cardinali
- Institute of Sciences of Food Productions (ISPA), National Research Council (CNR), 70126 Bari, Italy; (A.R.B.); (M.D.B.); (V.L.); (I.D.); (S.L.L.); (A.G.); (A.C.)
| |
Collapse
|
3
|
Salsinha AS, Cima A, Araújo-Rodrigues H, Viana S, Reis F, Coscueta ER, Rodríguez-Alcalá LM, Relvas JB, Pintado M. The use of an in vitro fecal fermentation model to uncover the beneficial role of omega-3 and punicic acid in gut microbiota alterations induced by a Western diet. Food Funct 2024; 15:6095-6117. [PMID: 38757812 DOI: 10.1039/d4fo00727a] [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: 05/18/2024]
Abstract
The influence of gut microbiota in the onset and development of several metabolic diseases has gained attention over the last few years. Diet plays an essential role in gut microbiota modulation. Western diet (WD), characterized by high-sugar and high-fat consumption, alters gut microbiome composition, diversity index, microbial relative levels, and functional pathways. Despite the promising health effects demonstrated by polyunsaturated fatty acids, their impact on gut microbiota is still overlooked. The effect of Fish oil (omega-3 source) and Pomegranate oil (punicic acid source), and a mixture of both oils in gut microbiota modulation were determined by subjecting the oil samples to in vitro fecal fermentations. Cecal samples from rats from two different dietary groups: a control diet (CD) and a high-fat high-sugar diet (WD), were used as fecal inoculum. 16S amplicon metagenomics sequencing showed that Fish oil + Pomegranate oil from the WD group increased α-diversity. This sample can also increase the relative abundance of the Firmicutes and Bacteroidetes phylum as well as Akkermansia and Blautia, which were affected by the WD consumption. All samples were able to increase butyrate and acetate concentration in the WD group. Moreover, tyrosine concentrations, a precursor for dopamine and norepinephrine, increase in the Fish oil + Pomegranate oil WD sample. GABA, an important neurotransmitter, was also increased in WD samples. These results suggest a potential positive impact of these oils' mixture on gut-brain axis modulation. It was demonstrated, for the first time, the great potential of using a mixture of both Fish and Pomegranate oil to restore the gut microbiota changes associated with WD consumption.
Collapse
Affiliation(s)
- Ana Sofia Salsinha
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal.
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto - Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - André Cima
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal.
| | - Helena Araújo-Rodrigues
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal.
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto - Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Sofia Viana
- Coimbra Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra, Coimbra, Portugal
- Instituto Politécnico de Coimbra, Escola Superior de Tecnologia da Saúde de Coimbra, Rua 5 de Outubro - S. Martinho Bispo, Apartado 7006, 3046-854 Coimbra, Portugal
| | - Flávio Reis
- Coimbra Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - Ezequiel R Coscueta
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal.
| | - Luis Miguel Rodríguez-Alcalá
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal.
| | - João B Relvas
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto - Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Departmento de Biomedicina, Faculdade de Medicina da Universidade do Porto (FMUP), 4200-319 Porto, Portugal
| | - Manuela Pintado
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal.
| |
Collapse
|
4
|
Rocha HR, Pintado ME, Gomes AM, Coelho MC. Carotenoids and Intestinal Harmony: Exploring the Link for Health. Foods 2024; 13:1599. [PMID: 38890828 PMCID: PMC11171705 DOI: 10.3390/foods13111599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/02/2024] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
Carotenoids, prominent lipid-soluble phytochemicals in the human diet, are responsible for vibrant colours in nature and play crucial roles in human health. While they are extensively studied for their antioxidant properties and contributions to vitamin A synthesis, their interactions with the intestinal microbiota (IM) remain poorly understood. In this study, beta (β)-carotene, lutein, lycopene, a mixture of these three pigments, and the alga Osmundea pinnatifida were submitted to simulated gastrointestinal digestion (GID) and evaluated on human faecal samples. The results showed varying effects on IM metabolic dynamics, organic acid production, and microbial composition. Carotenoid exposure influenced glucose metabolism and induced the production of organic acids, notably succinic and acetic acids, compared with the control. Microbial composition analysis revealed shifts in phyla abundance, particularly increased Pseudomonadota. The α-diversity indices demonstrated higher diversity in β-carotene and the pigments' mixture samples, while the β-diversity analysis indicated significant dissimilarity between the control and the carotenoid sample groups. UPLC-qTOF MS analysis suggested dynamic changes in carotenoid compounds during simulated fermentation, with lutein exhibiting distinct mass ion fragmentation patterns. This comprehensive research enhances our understanding of carotenoid-IM interactions, shedding light on potential health implications and the need for tailored interventions for optimal outcomes.
Collapse
Affiliation(s)
| | | | | | - Marta C. Coelho
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (H.R.R.); (M.E.P.); (A.M.G.)
| |
Collapse
|
5
|
Dixit Y, Kanojiya K, Bhingardeve N, Ahire JJ, Saroj D. In Vitro Human Gastrointestinal Tract Simulation Systems: A Panoramic Review. Probiotics Antimicrob Proteins 2024; 16:501-518. [PMID: 36988898 DOI: 10.1007/s12602-023-10052-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2023] [Indexed: 03/30/2023]
Abstract
Simulated human gastrointestinal (GI) tract systems are important for their applications in the fields of probiotics, nutrition and health. To date, various in vitro gut systems have been available to study GI tract dynamics and its association with health. In contrast to in vivo investigations, which are constrained by ethical considerations, in vitro models have several benefits despite the challenges involved in mimicking the GI environment. These in vitro models can be used for a range of research, from simple to dynamic, with one compartment to several compartments. In this review, we present a panoramic development of in vitro GI models for the first time through an evolutionary timeline. We tried to provide insight on designing an in vitro gut model, especially for novices. Latest developments and scope for improvement based on the limitations of the existing models were highlighted. In conclusion, designing an in vitro GI model suitable for a particular application is a multifaceted task. The bio-mimicking of the GI tract specific to geometrical, anatomical and mechanical features remains a challenge for the development of effective in vitro GI models. Advances in computer technology, artificial intelligence and nanotechnology are going to be revolutionary for further development. Besides this, in silico high-throughput technologies and miniaturisation are key players in the success of making in vitro modelling cost-effective and reducing the burden of in vivo studies.
Collapse
Affiliation(s)
- Yogini Dixit
- Advanced Enzyme Technologies Ltd., 5th Floor, A-Wing, Sun Magnetica, Louiswadi, Maharashtra, Thane West, India
| | - Khushboo Kanojiya
- Advanced Enzyme Technologies Ltd., 5th Floor, A-Wing, Sun Magnetica, Louiswadi, Maharashtra, Thane West, India
| | - Namrata Bhingardeve
- Advanced Enzyme Technologies Ltd., 5th Floor, A-Wing, Sun Magnetica, Louiswadi, Maharashtra, Thane West, India
| | - Jayesh J Ahire
- Advanced Enzyme Technologies Ltd., 5th Floor, A-Wing, Sun Magnetica, Louiswadi, Maharashtra, Thane West, India.
| | - Dina Saroj
- Advanced Enzyme Technologies Ltd., 5th Floor, A-Wing, Sun Magnetica, Louiswadi, Maharashtra, Thane West, India
| |
Collapse
|
6
|
Kozak T, Lykhova O, Serhiichuk T, Bezdieniezhnykh N, Chekhun V. OPTIMIZATION OF EXPERIMENTAL MODEL SYSTEMS FOR EVALUATING RECIPROCAL INFLUENCE OF BIFIDOBACTERIUM ANIMALIS AND HUMAN BREAST CANCER CELLS IN VITRO. Exp Oncol 2024; 45:504-514. [PMID: 38328839 DOI: 10.15407/exp-oncology.2023.04.504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Indexed: 02/09/2024]
Abstract
BACKGROUND The development of human breast cancer (BC) is known to be closely related to disturbances in the mammary gland microbiota. Bacteria of the genus Bifidobacterium are an important component of normal breast microbiota and exert antitumor activity. The molecular-biological mechanisms of interaction between BC cells and microbiota members remain poorly studied yet. The aim of this study was to develop and optimize an experimental model system for the co-cultivation of BC cells with Bifidobacterium animalis in vitro. MATERIALS AND METHODS Human ВС cells of the MCF-7, T47D, and MDA-MB-231 lines, as well as live and heat-inactivated bacteria of Bifidobacterium animalis subsp. lactis (B. animalis) were used as research objects. The growth kinetics and viability of B. animalis in the presence of different ВС cell lines and without them were determined by both the turbidimetry method and seeding on an elective nutrient medium. Glucose consumption and lactate production by bifidobacteria were assessed by biochemical methods. The viability of BC cells was determined by a standard colorimetric method. RESULTS The growth kinetics of B. animalis in the complete DMEM nutrient medium showed standard patterns. The indicators of glucose consumption and lactate production of B. animalis confirm its physiological metabolic activity under the growth conditions. The presence of BC cells in the model system did not affect the duration of the growth phases of the B. animalis cells' population but contributed to the increase in their counts. A significant decrease in the number of live BC cells of all studied lines was observed only after 48 h of co-cultivation with live B. animalis. To achieve similar suppression of the BC cell viability, 10-30-fold higher counts of heatinactivated bacteria were required compared to live ones. CONCLUSIONS The optimal conditions for co-cultivation of human BC cells and living B. animalis cells in vitro have been identified.
Collapse
Affiliation(s)
- T Kozak
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, Kyiv, Ukraine
| | - O Lykhova
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, Kyiv, Ukraine
| | - T Serhiichuk
- Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - N Bezdieniezhnykh
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, Kyiv, Ukraine
| | - V Chekhun
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, Kyiv, Ukraine
| |
Collapse
|
7
|
Rachmühl C, Lacroix C, Cabrera PM, Geirnaert A. Long-term continuous cultivation of Kenyan infant fecal microbiota using the host adapted PolyFermS model. Sci Rep 2023; 13:20563. [PMID: 37996456 PMCID: PMC10667343 DOI: 10.1038/s41598-023-47131-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023] Open
Abstract
Appropriate in vitro models to investigate the impact of novel nutritional strategies on the gut microbiota of infants living in rural Africa are scarce. Here, we aimed to develop such a continuous gut fermentation model based on the PolyFermS platform, which allows controlled and stable long-term cultivation of colon microbiota in conditions akin the host. Nine immobilized Kenyan infant fecal microbiota were used as inoculum for continuous PolyFermS colon models fed with medium mimicking the weaning infant diet. Fructo-oligosaccharides (FOS) supplementation (1, 4 and 8 g/L) and cultivation pH (5.8 and 6.3) were investigated stepwise. Conditions providing a close match between fecal and in vitro microbiota (pH 5.8 with 1 g/L FOS) were selected for investigating long-term stability of four Kenyan infant PolyFermS microbiota. The shared fraction of top bacterial genera between fecal and in vitro microbiota was high (74-89%) and stable during 107 days of continuous cultivation. Community diversity was maintained and two distinct fermentation metabolite profiles of infant fecal microbiota were observed. Three propiogenic and one butyrogenic metabolite profile of infant fecal microbiota established from day 8 onwards and stayed stable. We present here the first rationally designed continuous cultivation model of African infant gut microbiota. This model will be important to assess the effect of dietary or environmental factors on the gut microbiota of African infants with high enteropathogen exposure.
Collapse
Affiliation(s)
- Carole Rachmühl
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland.
| | - Paula Momo Cabrera
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Annelies Geirnaert
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
8
|
Zhang T, Liu W, Lu H, Cheng T, Wang L, Wang G, Zhang H, Chen W. Lactic acid bacteria in relieving constipation: mechanism, clinical application, challenge, and opportunity. Crit Rev Food Sci Nutr 2023; 65:551-574. [PMID: 37971876 DOI: 10.1080/10408398.2023.2278155] [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: 11/19/2023]
Abstract
Constipation is a prevalent gastrointestinal symptom that can considerably affect a patients' quality of life. Although several drugs have been used to treat constipation, they are associated with high costs, side effects, and low universality. Therefore, alternative intervention strategies are urgently needed. Traditional lactic acid bacteria (LAB), such as Bifidobacterium and Lactobacillus, play a vital role in regulating intestinal microecology and have demonstrated favorable effects in constipation; however, a comprehensive review of their constipation relief mechanisms is limited. This review summarizes the pathogenesis of constipation and the relationship between intestinal motility and gut microbiota, elucidates the possible mechanism by which LAB alleviates of constipation through a systematic summary of animal and clinical research, and highlights the challenges and applications of LAB in the treatment of constipation. Our review can improve our understanding of constipation, and advance targeted microecological therapeutic agents, such as LAB.
Collapse
Affiliation(s)
- Tong Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wenxu Liu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Huimin Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Ting Cheng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Linlin Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, China
| | - Gang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| |
Collapse
|
9
|
Lai Y, Deng H, Fang Q, Ma L, Lei H, Guo X, Chen Y, Song C. Water-Insoluble Polysaccharide Extracted from Poria cocos Alleviates Antibiotic-Associated Diarrhea Based on Regulating the Gut Microbiota in Mice. Foods 2023; 12:3080. [PMID: 37628079 PMCID: PMC10453245 DOI: 10.3390/foods12163080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/03/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Antibiotics are very effective in treating a variety of bacterial infections, while clinical overuse of antibiotics can lead to diseases such as antibiotic-associated diarrhea. Numerous studies have shown that natural polysaccharides can be used as prebiotics to alleviate antibiotic-associated diarrhea (AAD). Poria cocos is a medicinal and edible mushroom widely used for thousands of years in China, and our former study demonstrated that water-insoluble polysaccharide (PCY) has the potential prebiotic function. Therefore, we simulated the digestion and fermentation of PCY using feces from volunteers, and then administered it to C57BL/6 mice with AAD to study its effects on the gut microbiota and metabolites. The results indicated that PCY effectively alleviated the symptoms of AAD in mice, restored the intestinal barrier function, improved the content of short-chain fatty acids (SCFAs), decreased the level of inflammatory cytokines, and changed the structure of gut microbiota by increasing the relative abundance of norank_f__Muribaculaceae and unclassified_f__Lachnospiraceae, and decreasing that of Escherichia-Shigella, Staphylococcus and Acinetobacter. This study further demonstrated that PCY is an effective functional prebiotic for improving AAD disease, and provided a new avenue and insight for developing PCY as a functional food or prebiotic for alleviating gastrointestinal diseases.
Collapse
Affiliation(s)
- Yong Lai
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (Y.L.); (Q.F.); (L.M.); (H.L.); (X.G.)
| | - Huiling Deng
- Chongqing Academy of Science and Technology, Chongqing 401121, China; (H.D.); (Y.C.)
- Key Laboratory of Condiment Supervision Technology for State Market Regulation, Chongqing Institute for Food and Drug Administration, Chongqing 401121, China
| | - Qi Fang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (Y.L.); (Q.F.); (L.M.); (H.L.); (X.G.)
| | - Linhua Ma
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (Y.L.); (Q.F.); (L.M.); (H.L.); (X.G.)
| | - Hui Lei
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (Y.L.); (Q.F.); (L.M.); (H.L.); (X.G.)
| | - Xiurong Guo
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (Y.L.); (Q.F.); (L.M.); (H.L.); (X.G.)
| | - Ya Chen
- Chongqing Academy of Science and Technology, Chongqing 401121, China; (H.D.); (Y.C.)
| | - Can Song
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (Y.L.); (Q.F.); (L.M.); (H.L.); (X.G.)
| |
Collapse
|
10
|
Rachmühl C, Lacroix C, Cabrera PM, Geirnaert A. Long-term continuous cultivation of Kenyan infant fecal microbiota using the host adapted PolyFermS model. RESEARCH SQUARE 2023:rs.3.rs-3101157. [PMID: 37461546 PMCID: PMC10350169 DOI: 10.21203/rs.3.rs-3101157/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Appropriate in vitro models to investigate the impact of novel nutritional strategies on the gut microbiota of infants living in rural Africa are scarce. Here, we aimed to develop such a continuous gut fermentation model based on the PolyFermS platform. Eight immobilized Kenyan infant fecal microbiota were used as inoculum for continuous PolyFermS colon models fed with medium mimicking the weaning infant diet. Fructo-oligosaccharides (FOS) supplementation (1, 4 and 8 g/L) and cultivation pH (5.8 and 6.3) were stepwise investigated. Conditions providing a close match between fecal and in vitro microbiota (pH 5.8 with 1 g/L FOS) were selected for investigating long-term stability of four Kenyan infant PolyFermS microbiota. The shared fraction of top bacterial genera between fecal and in vitro microbiota was high (74-89%) and stable during 107 days of continuous cultivation. Community diversity was maintained, and two distinct fermentation metabolite profiles, propiogenic and butyrogenic, of infant fecal microbiota established from day 8 onwards and stayed stable. We present here the first rationally designed and accurate continuous cultivation model of African infant gut microbiota. This model will be important to assess the effect of dietary or environmental factors on the gut microbiota of African infants with high enteropathogen exposure.
Collapse
|
11
|
Liu M, Liu Z, Zhang N, Cao Z, Fu J, Yuan W, Wu H, Shang H. Preparation of polysaccharides from Crepis tectorum Linn. and the regulation effects on intestinal microbiota. Process Biochem 2023. [DOI: 10.1016/j.procbio.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
12
|
Gonçalves DA, González A, Roupar D, Teixeira JA, Nobre C. How prebiotics have been produced from agro-industrial waste: An overview of the enzymatic technologies applied and the models used to validate their health claims. Trends Food Sci Technol 2023. [DOI: 10.1016/j.tifs.2023.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
13
|
Rehman A, Pham V, Seifert N, Richard N, Sybesma W, Steinert RE. The Polyunsaturated Fatty Acids Eicosapentaenoic Acid and Docosahexaenoic Acid, and Vitamin K 1 Modulate the Gut Microbiome: A Study Using an In Vitro Shime Model. J Diet Suppl 2023; 21:135-153. [PMID: 37078491 DOI: 10.1080/19390211.2023.2198007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
Omega-3 polyunsaturated fatty acids (PUFAs) and vitamins exert multiple beneficial effects on host health, some of which may be mediated through the gut microbiome. We investigated the prebiotic potential of eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA) and lipid-soluble phylloquinone (vitamin K1), each at 0.2x, 1x and 5x using the simulator of the human intestinal microbial ecosystem (SHIME®) to exclude in vivo systemic effects and host-microbe interactions.Microbial community composition and, diversity [shotgun metagenomic sequencing] and microbial activity [pH, gas pressure, and production of short-chain fatty acids (SCFAs)] were measured over a period of 48 h. Fermentations supernatants were used to investigate the effect on gut barrier integrity using a Caco-2/goblet cell co-culture model.We found that EPA, DHA and vitamin K1 increased alpha-diversity at 24 h when compared with control. Moreover, there was an effect on beta-diversity with changes in gut microbial composition, such as an increase in the Firmicutes/Bacteroidetes (F/B) ratio and a consistent increase in Veillonella and Dialister abundances with all treatments. DHA, EPA, and vitamin K1 also modulated metabolic activity of the gut microbiome by increasing total SCFAs which was related mainly to an increase in propionate (highest with EPA and vitamin K1 at 0.2x). Finally, we found that EPA and DHA increased gut barrier integrity with DHA at 1x and EPA at 5x (p < 0.05, respectively). In conclusion, our in vitro data further establish a role of PUFAs and vitamin K to modulate the gut microbiome with effects on the production of SCFAs and barrier integrity.
Collapse
Affiliation(s)
- Ateequr Rehman
- Human Nutrition and Health, DSM Nutritional Products Ltd, Basel, Switzerland
| | - Van Pham
- Human Nutrition and Health, DSM Nutritional Products Ltd, Basel, Switzerland
| | - Nicole Seifert
- Human Nutrition and Health, DSM Nutritional Products Ltd, Basel, Switzerland
| | - Nathalie Richard
- Human Nutrition and Health, DSM Nutritional Products Ltd, Basel, Switzerland
| | - Wilbert Sybesma
- Human Nutrition and Health, DSM Nutritional Products Ltd, Basel, Switzerland
| | - Robert E Steinert
- Human Nutrition and Health, DSM Nutritional Products Ltd, Basel, Switzerland
- Department of Surgery, Division of Visceral and Transplantation Surgery, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
14
|
An In Vitro Model of the Chicken Gastrointestinal Tract with Special Emphasis to the Cecal Microbiota. Poult Sci 2023; 102:102654. [PMID: 37043954 PMCID: PMC10140163 DOI: 10.1016/j.psj.2023.102654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
An in vitro model of the upper gastrointestinal tract as well as the chicken cecum was developed to have a predictive tool for estimating the production performance of animals by analyzing the feeding value of a certain diet. The upper gastrointestinal tract consists of a batch type model, whereas the cecal model is comprised of 4 semi-continuous connected vessels inoculated with cecal or fecal microbes. The upper gastrointestinal tract and cecal simulations were both run with a corn- and a wheat-based diet to simulate 2 typical feed types. Samples were collected after the 5-h cecal simulations and aliquots were frozen to assess inoculum stability. The microbiota was analyzed by 16S rRNA gene sequencing, whereas short chain fatty acids as microbial metabolites were analyzed by using gas chromatography. As expected, some significant differences in microbial abundance after simulation between the cecal and fecal slurry samples (P = 0.001) were detected, as well between the fresh and frozen status (P = 0.001), hence simulations inoculated with cecal and fresh samples being more diverse. For the measured metabolites, almost all of them increased (P < 0.05) significantly when comparing fresh and frozen inoculum. The present chicken intestinal in vitro model represents a rapid systematic screening system for studying dietary related microbial changes and reducing the need of animal sacrifice for experimentation.
Collapse
|
15
|
Tian J, Wang X, Zhang X, Chen X, Dong M, Rui X, Zhang Q, Jiang M, Li W. Artificial simulated saliva, gastric and intestinal digestion and fermentation in vitro by human gut microbiota of intrapolysaccharide from Paecilomyces cicadae TJJ1213. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2022.07.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
16
|
Lee KW, Shin JS, Lee CM, Han HY, O Y, Kim HW, Cho TJ. Gut-on-a-Chip for the Analysis of Bacteria-Bacteria Interactions in Gut Microbial Community: What Would Be Needed for Bacterial Co-Culture Study to Explore the Diet-Microbiota Relationship? Nutrients 2023; 15:nu15051131. [PMID: 36904133 PMCID: PMC10005057 DOI: 10.3390/nu15051131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 02/26/2023] Open
Abstract
Bacterial co-culture studies using synthetic gut microbiomes have reported novel research designs to understand the underlying role of bacterial interaction in the metabolism of dietary resources and community assembly of complex microflora. Since lab-on-a-chip mimicking the gut (hereafter "gut-on-a-chip") is one of the most advanced platforms for the simulative research regarding the correlation between host health and microbiota, the co-culture of the synthetic bacterial community in gut-on-a-chip is expected to reveal the diet-microbiota relationship. This critical review analyzed recent research on bacterial co-culture with perspectives on the ecological niche of commensals, probiotics, and pathogens to categorize the experimental approaches for diet-mediated management of gut health as the compositional and/or metabolic modulation of the microbiota and the control of pathogens. Meanwhile, the aim of previous research on bacterial culture in gut-on-a-chip has been mainly limited to the maintenance of the viability of host cells. Thus, the integration of study designs established for the co-culture of synthetic gut consortia with various nutritional resources into gut-on-a-chip is expected to reveal bacterial interspecies interactions related to specific dietary patterns. This critical review suggests novel research topics for co-culturing bacterial communities in gut-on-a-chip to realize an ideal experimental platform mimicking a complex intestinal environment.
Collapse
Affiliation(s)
- Ki Won Lee
- Department of Food and Biotechnology, College of Science and Technology, Korea University, 2511, Sejong-ro, Sejong 30019, Republic of Korea
| | - Jin Song Shin
- Department of Food Regulatory Science, College of Science and Technology, Korea University, 2511, Sejong-ro, Sejong 30019, Republic of Korea
| | - Chan Min Lee
- Department of Food and Biotechnology, College of Science and Technology, Korea University, 2511, Sejong-ro, Sejong 30019, Republic of Korea
| | - Hea Yeon Han
- Department of Food and Biotechnology, College of Science and Technology, Korea University, 2511, Sejong-ro, Sejong 30019, Republic of Korea
| | - Yun O
- Department of Food Regulatory Science, College of Science and Technology, Korea University, 2511, Sejong-ro, Sejong 30019, Republic of Korea
| | - Hye Won Kim
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Tae Jin Cho
- Department of Food and Biotechnology, College of Science and Technology, Korea University, 2511, Sejong-ro, Sejong 30019, Republic of Korea
- Department of Food Regulatory Science, College of Science and Technology, Korea University, 2511, Sejong-ro, Sejong 30019, Republic of Korea
- Correspondence: ; Tel.: +82-44-860-1433
| |
Collapse
|
17
|
Średnicka P, Roszko MŁ, Popowski D, Kowalczyk M, Wójcicki M, Emanowicz P, Szczepańska M, Kotyrba D, Juszczuk-Kubiak E. Effect of in vitro cultivation on human gut microbiota composition using 16S rDNA amplicon sequencing and metabolomics approach. Sci Rep 2023; 13:3026. [PMID: 36810418 PMCID: PMC9945476 DOI: 10.1038/s41598-023-29637-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/08/2023] [Indexed: 02/23/2023] Open
Abstract
Gut microbiota (GM) plays many key functions and helps maintain the host's health. Consequently, the development of GM cultivation under in vitro stimulating physiological conditions has gained extreme interest in different fields. In this study, we evaluated the impact of four culture media: Gut Microbiota Medium (GMM), Schaedler Broth (SM), Fermentation Medium (FM), and Carbohydrate Free Basal Medium (CFBM) on preserving the biodiversity and metabolic activity of human GM in batch in vitro cultures using PMA treatment coupled with 16S rDNA sequencing (PMA-seq) and LC-HR-MS/MS untargeted metabolomics supplemented with GC-MS SCFA profiling. Before the experiments, we determined the possibility of using the pooled faecal samples (MIX) from healthy donors (n = 15) as inoculum to reduce the number of variables and ensure the reproducibility of in vitro cultivation tests. Results showed the suitability of pooling faecal samples for in vitro cultivation study. Non-cultured MIX inoculum was characterized by higher α-diversity (Shannon effective count, and Effective microbial richness) compared to inocula from individual donors. After 24 h of cultivation, a significant effect of culture media composition on GM taxonomic and metabolomic profiles was observed. The SM and GMM had the highest α-diversity (Shannon effective count). The highest number of core ASVs (125) shared with non-cultured MIX inoculum and total SCFAs production was observed in the SM. These results might contribute to the development of standardized protocols for human GM in vitro cultivation by preventing methodological bias in the data.
Collapse
Affiliation(s)
- Paulina Średnicka
- Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology - State Research Institute, Rakowiecka 36 Street, 02-532, Warsaw, Poland
| | - Marek Łukasz Roszko
- Department of Food Safety and Chemical Analysis, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology - State Research Institute, Rakowiecka 36 Street, 02-532, Warsaw, Poland.
| | - Dominik Popowski
- Department of Food Safety and Chemical Analysis, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology - State Research Institute, Rakowiecka 36 Street, 02-532, Warsaw, Poland
- Microbiota Lab, Department of Pharmacognosy and Molecular Basis of Phytotherapy, Medical University of Warsaw, Banacha 1 Street, 02-097, Warsaw, Poland
| | - Monika Kowalczyk
- Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology - State Research Institute, Rakowiecka 36 Street, 02-532, Warsaw, Poland
| | - Michał Wójcicki
- Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology - State Research Institute, Rakowiecka 36 Street, 02-532, Warsaw, Poland
| | - Paulina Emanowicz
- Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology - State Research Institute, Rakowiecka 36 Street, 02-532, Warsaw, Poland
| | - Magdalena Szczepańska
- Department of Food Safety and Chemical Analysis, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology - State Research Institute, Rakowiecka 36 Street, 02-532, Warsaw, Poland
| | - Danuta Kotyrba
- Department of Research, Scientific Information and Marketing Coordination, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology - State Research Institute, Rakowiecka 36 Street, 02-532, Warsaw, Poland
| | - Edyta Juszczuk-Kubiak
- Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology - State Research Institute, Rakowiecka 36 Street, 02-532, Warsaw, Poland.
| |
Collapse
|
18
|
Chen J, Yuan Z, Tu Y, Hu W, Xie C, Ye L. Experimental and computational models to investigate intestinal drug permeability and metabolism. Xenobiotica 2023; 53:25-45. [PMID: 36779684 DOI: 10.1080/00498254.2023.2180454] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Oral administration is the preferred route for drug administration that leads to better therapy compliance. The intestine plays a key role in the absorption and metabolism of oral drugs, therefore, new intestinal models are being continuously proposed, which contribute to the study of intestinal physiology, drug screening, drug side effects, and drug-drug interactions.Advances in pharmaceutical processes have produced more drug formulations, causing challenges for intestinal models. To adapt to the rapid evolution of pharmaceuticals, more intestinal models have been created. However, because of the complexity of the intestine, few models can take all aspects of the intestine into account, and some functions must be sacrificed to investigate other areas. Therefore, investigators need to choose appropriate models according to the experimental stage and other requirements to obtain the desired results.To help researchers achieve this goal, this review summarised the advantages and disadvantages of current commonly used intestinal models and discusses possible future directions, providing a better understanding of intestinal models.
Collapse
Affiliation(s)
- Jinyuan Chen
- Institute of Scientific Research, Southern Medical University, Guangzhou, P.R. China.,TCM-Integrated Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Ziyun Yuan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Yifan Tu
- Boehringer-Ingelheim, Connecticut, P.R. USA
| | - Wanyu Hu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Cong Xie
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Ling Ye
- TCM-Integrated Hospital, Southern Medical University, Guangzhou, P.R. China
| |
Collapse
|
19
|
In Vitro Fermentation of Edible Mushrooms: Effects on Faecal Microbiota Characteristics of Autistic and Neurotypical Children. Microorganisms 2023; 11:microorganisms11020414. [PMID: 36838379 PMCID: PMC9959845 DOI: 10.3390/microorganisms11020414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Children with autism spectrum disorder (ASD) often suffer gastrointestinal disturbances consistent with gut microbiota (GM) alterations. Treatment with pro/prebiotics may potentially alleviate gut symptoms, but the evidence for prebiotics is scarce. This study aims to evaluate the effects of edible mushrooms (Pleurotus, Basidiomycota) and prebiotic compounds on GM composition and metabolite production in vitro, using faecal samples from autistic and non-autistic children. Specific microbial populations were enumerated after 24 h of fermentation by quantitative PCR, and the metabolic production was determined by gas chromatography. Higher levels of Prevotella spp. and Bifidobacterium spp. were measured in neurotypical children compared to ASD children. A total of 24 h fermentation of Pleurotus eryngii and P. ostreatus mushroom powder increased the levels of Bifidobacterium, while known prebiotics increased the levels of total bacteria and Bacteroides in both groups. Only P. eryngii mushrooms resulted in significantly elevated levels of total bacteria Bacteroides and Feacalibacterium prausnitzii compared to the negative control (NC) in the ASD group. Both mushrooms induced elevated levels of butyrate after 24 h of fermentation, while short-chain fructooligosaccharides induced increased levels of acetate in the ASD group, compared to NC. Overall, this study highlights the positive effect of edible mushrooms on the GM and metabolic activity of children with ASD.
Collapse
|
20
|
Isenring J, Bircher L, Geirnaert A, Lacroix C. In vitro human gut microbiota fermentation models: opportunities, challenges, and pitfalls. MICROBIOME RESEARCH REPORTS 2023; 2:2. [PMID: 38045607 PMCID: PMC10688811 DOI: 10.20517/mrr.2022.15] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/12/2022] [Accepted: 01/09/2023] [Indexed: 12/05/2023]
Abstract
The human gut microbiota (HGM) plays a pivotal role in health and disease. Consequently, nutritional and medical research focusing on HGM modulation strategies as a means of improving host health is steadily increasing. In vitro HGM fermentation models offer a valid complement to human and animal studies when it comes to the mechanistic exploration of novel modulation approaches and their direct effects on HGM composition and activity, while excluding interfering host effects. However, in vitro cultivation of HGM can be challenging due to its high oxygen sensitivity and the difficulties of accurately modeling the physio-chemical complexity of the gut environment. Despite the increased use of in vitro HGM models, there is no consensus about appropriate model selection and operation, sometimes leading to major deficiencies in study design and result interpretation. In this review paper, we aim to analyze crucial aspects of the application, setup and operation, data validation and result interpretation of in vitro HGM models. When carefully designed and implemented, in vitro HGM modeling is a powerful strategy for isolating and investigating biotic and abiotic factors in the HGM, as well as evaluating their effects in a controlled environment akin to the gut. Furthermore, complementary approaches combining different in vitro and in vivo models can strengthen the design and interpretation of human studies.
Collapse
Affiliation(s)
| | | | | | - Christophe Lacroix
- Department of Health Sciences and Technology, ETH Zurich, Zürich 8092, Switzerland
| |
Collapse
|
21
|
Ewin D, Birch WD, Moura IB. In vitro models to study Clostridioides difficile infection: current systems and future advances. Curr Opin Gastroenterol 2023; 39:23-30. [PMID: 36504033 PMCID: PMC9799034 DOI: 10.1097/mog.0000000000000893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Clostridioides difficile infection (CDI) is the most common cause of healthcare-associated diarrhoea in western countries, being categorized as an urgent healthcare threat. Historically, researchers have relied on the use of in vivo animal models to study CDI pathogenesis; however, differences in physiology and disease prognosis compared with humans limit their suitability to model CDI. In vitro models are increasingly being used as an alternative as they offer excellent process control, and some are able to use human ex-vivo prokaryotic and/or eukaryotic cells. RECENT FINDINGS Simulating the colonic environment in vitro is particularly challenging. Bacterial fermentation models have been used to evaluate novel therapeutics, explore the re-modelling of the gut microbiota, and simulate disease progression. However, they lack the scalability to become more widespread. Models that co-culture human and bacterial cells are of particular interest, but the different conditions required by each cell type make these models challenging to run. Recent advancements in model design have allowed for longer culture times with more representative bacterial populations. SUMMARY As in vitro models continue to evolve, they become more physiologically relevant, offering improved simulations of CDI, and extending their applicability.
Collapse
Affiliation(s)
- Duncan Ewin
- Healthcare-Associated Infections Group, Leeds Institute of Medical Research, Faculty of Medicine and Health
| | | | - Ines B. Moura
- Healthcare-Associated Infections Group, Leeds Institute of Medical Research, Faculty of Medicine and Health
| |
Collapse
|
22
|
Refael G, Riess HT, Levi CS, Magzal F, Tamir S, Koren O, Lesmes U. Responses of the human gut microbiota to physiologically digested insect powders or isolated chitin thereof. FUTURE FOODS 2022. [DOI: 10.1016/j.fufo.2022.100197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
23
|
Exploring species-level infant gut bacterial biodiversity by meta-analysis and formulation of an optimized cultivation medium. NPJ Biofilms Microbiomes 2022; 8:88. [PMID: 36316342 PMCID: PMC9622858 DOI: 10.1038/s41522-022-00349-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
In vitro gut cultivation models provide host-uncoupled, fast, and cost-efficient solutions to investigate the effects of intrinsic and extrinsic factors impacting on both composition and functionality of the intestinal microbial ecosystem. However, to ensure the maintenance and survival of gut microbial players and preserve their functions, these systems require close monitoring of several variables, including oxygen concentration, pH, and temperature, as well as the use of a culture medium satisfying the microbial nutritional requirements. In this context, in order to identify the macro- and micro-nutrients necessary for in vitro cultivation of the infant gut microbiota, a meta-analysis based on 1669 publicly available shotgun metagenomic samples corresponding to fecal samples of healthy, full-term infants aged from a few days to three years was performed to define the predominant species characterizing the “infant-like” gut microbial ecosystem. A subsequent comparison of growth performances was made using infant fecal samples that contained the most abundant bacterial taxa of the infant gut microbiota, when cultivated on 18 different culture media. This growth analysis was performed by means of flow cytometry-based bacterial cell enumeration and shallow shotgun sequencing, which allowed the formulation of an optimized growth medium, i.e., Infant Gut Super Medium (IGSM), which maintains and sustains the infant gut microbial biodiversity under in vitro growth conditions. Furthermore, this formulation was used to evaluate the in vitro effect of two drugs commonly used in pediatrics, i.e., acetaminophen and simethicone, on the taxonomic composition of the infant gut microbiota.
Collapse
|
24
|
Singh V, Son H, Lee G, Lee S, Unno T, Shin JH. Role, Relevance, and Possibilities of In vitro fermentation models in human dietary, and gut-microbial studies. Biotechnol Bioeng 2022; 119:3044-3061. [PMID: 35941765 DOI: 10.1002/bit.28206] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/12/2022] [Accepted: 08/03/2022] [Indexed: 11/11/2022]
Abstract
Dietary studies play a crucial role in determining the health-benefiting effects of most food substances, including prebiotics, probiotics, functional foods, and bioactive compounds. Such studies involve gastrointestinal digestion and colonic fermentation of dietary substances. In colonic fermentation, any digested food is further metabolized in the gut by the residing colonic microbiota, causing a shift in the gut microenvironment and production of various metabolites, such as short-chain fatty acids (SCFA). These diet-induced shifts in the microbial community and metabolite production, which can be assessed through in vitro fermentation models using a donor's fecal microbiota, are well known to impact the health of the host. Although in vivo or animal experiments are the gold standard in dietary studies, recent advancements using different in vitro systems, like artificial colon (ARCOL), mini bioreactor array (MBRA), TNO in vitro model of the colon (TIM), Simulator of the Human Intestinal Microbial Ecosystem (SHIME), M-SHIME, CoMiniGut, and Dynamic Gastrointestinal Simulator (SIMGI) make it easy to study the dietary impact in terms of the gut microbiota and metabolites. Such a continuous in vitro system can have multiple compartments corresponding to different parts of the colon, i.e., proximal, transverse, and distal colon, making the findings physiologically more significant. Further, post-fermentation samples can be analyzed using metagenomic, metabolomic, qPCR and flow cytometry approaches. Moreover, studies have shown that in vitro results are in accordance with the in vivo findings, supporting their relevance in dietary studies and giving confidence that shifts in metabolites are only due to microbes. This review meticulously describes the recent advancements in various fermentation models and their relevance in dietary studies. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Vineet Singh
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| | - HyunWoo Son
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| | - GyuDae Lee
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| | - Sunwoo Lee
- Department of Biotechnology,, School of Life Sciences, SARI, Jeju National University, Jeju, South Korea
| | - Tatsuya Unno
- Department of Biotechnology,, School of Life Sciences, SARI, Jeju National University, Jeju, South Korea
| | - Jae-Ho Shin
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
25
|
Pi X, Yu Z, Yang X, Du Z, Liu W. Effects of Zymosan on Short-Chain Fatty Acid and Gas Production in in vitro Fermentation Models of the Human Intestinal Microbiota. Front Nutr 2022; 9:921137. [PMID: 35859755 PMCID: PMC9291218 DOI: 10.3389/fnut.2022.921137] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/06/2022] [Indexed: 11/27/2022] Open
Abstract
In this study, the effects of zymosan (HG, hydrolyzed glucan) on the structure and metabolism of fecal microbiota in Chinese healthy people was investigated by an in vitro simulated intestinal microecology fermentation model. We found that HG significantly regulated fecal microbiota composition, including the increase of Bifidobacterium, Faecalibacterium, Prevotella and the decrease of Escherichia-Shigella. Moreover, HG significantly increased the total production of short chain fatty acids (SCFAs) and gases, in which the production of Acetic acid, Propionic acid, CO2, and H2 significantly increased while the production of Isovaleric acid and NH3 significantly decreased. Additionally, the supplement of HG showed certain differences in the regulation of microbiota from four groups. HG significantly increased the relative abundance of Bifidobacterium and significantly decreased the relative abundance of Escherichia-Shigella excluding the older men group. Meanwhile, and the relative abundance of Lactobacillus was significantly increased in young populations. And the relative abundance of Bacteroides was significantly decreased only in the young women. Furthermore, HG significantly increased H2 concentration only in older men. These findings suggest that HG, as a new generation of prebiotics, could regulate the structure of fecal microbiota and its metabolites in a better direction, but when HG participates in precision nutrition formula, it may be necessary to consider the differences in the utilization of different populations.
Collapse
Affiliation(s)
- Xionge Pi
- Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Zaichun Yu
- College of Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Xiaoxia Yang
- College of Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Zhi Du
- Department of Pharmacy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Wei Liu
- Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
- *Correspondence: Wei Liu
| |
Collapse
|
26
|
Hobson CA, Vigue L, Naimi S, Chassaing B, Magnan M, Bonacorsi S, Gachet B, El Meouche I, Birgy A, Tenaillon O. MiniBioReactor Array (MBRA) in vitro gut model: a reliable system to study microbiota-dependent response to antibiotic treatment. JAC Antimicrob Resist 2022; 4:dlac077. [PMID: 35795241 PMCID: PMC9252984 DOI: 10.1093/jacamr/dlac077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 06/14/2022] [Indexed: 11/18/2022] Open
Abstract
Background Antimicrobial drugs are mostly studied for their impact on emergence of bacterial antibiotic resistance, but their impact on the gut microbiota is also of tremendous interest. In vitro gut models are important tools to study such complex drug–microbiota interactions in humans. Methods The MiniBioReactor Array (MBRA) in vitro microbiota system; a single-stage continuous flow culture model, hosted in an anaerobic chamber; was used to evaluate the impact of three concentrations of a third-generation cephalosporin (ceftriaxone) on faecal microbiota from two healthy donors (treatment versus control: three replicates per condition). We conducted 16S microbiome profiling and analysed microbial richness, diversity and taxonomic changes. β-Lactamase activities were evaluated and correlated with the effects observed in the MBRA in vitro system. Results The MBRA preserved each donor’s specificities, and differences between the donors were maintained through time. Before treatment, all faecal cultures belonging to the same donor were comparable in composition, richness, and diversity. Treatment with ceftriaxone was associated with a decrease in α-diversity, and an increase in β-diversity index, in a concentration-dependent manner. The maximum effect on diversity was observed after 72 h of treatment. Importantly, one donor had a stronger microbiota β-lactamase activity that was associated with a reduced impact of ceftriaxone on microbiota composition. Conclusions MBRA can reliably mimic the intestinal microbiota and its modifications under antibiotic selective pressure. The impact of the treatment was donor- and concentration-dependent. We hypothesize these results could be explained, at least in part, by the differences in β-lactamase activity of the microbiota itself. Our results support the relevance and promise of the MBRA system to study drug–microbiota interactions.
Collapse
Affiliation(s)
- C A Hobson
- IAME, UMR 1137, INSERM, Université de Paris, AP-HP , Paris , France
| | - L Vigue
- IAME, UMR 1137, INSERM, Université de Paris, AP-HP , Paris , France
| | - S Naimi
- INSERM U1016, Team ‘Mucosal Microbiota in Chronic Inflammatory diseases’, CNRS UMR 8104, Université de Paris , Paris , France
| | - B Chassaing
- INSERM U1016, Team ‘Mucosal Microbiota in Chronic Inflammatory diseases’, CNRS UMR 8104, Université de Paris , Paris , France
| | - M Magnan
- IAME, UMR 1137, INSERM, Université de Paris, AP-HP , Paris , France
| | - S Bonacorsi
- IAME, UMR 1137, INSERM, Université de Paris, AP-HP , Paris , France
- Laboratoire de Microbiologie, Hôpital Robert Debré, AP-HP , 75019 Paris , France
| | - B Gachet
- IAME, UMR 1137, INSERM, Université de Paris, AP-HP , Paris , France
| | - I El Meouche
- IAME, UMR 1137, INSERM, Université de Paris, AP-HP , Paris , France
| | - A Birgy
- IAME, UMR 1137, INSERM, Université de Paris, AP-HP , Paris , France
- Laboratoire de Microbiologie, Hôpital Robert Debré, AP-HP , 75019 Paris , France
| | - O Tenaillon
- IAME, UMR 1137, INSERM, Université de Paris, AP-HP , Paris , France
| |
Collapse
|
27
|
Ahire J, Mokashe N, Kashikar M, Madempudi R. Survival of Limosilactobacillus reuteri UBLRu-87 during passage through the in vitro gut model system. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2022.113652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
28
|
Gibbons SM, Gurry T, Lampe JW, Chakrabarti A, Dam V, Everard A, Goas A, Gross G, Kleerebezem M, Lane J, Maukonen J, Penna ALB, Pot B, Valdes AM, Walton G, Weiss A, Zanzer YC, Venlet NV, Miani M. Perspective: Leveraging the Gut Microbiota to Predict Personalized Responses to Dietary, Prebiotic, and Probiotic Interventions. Adv Nutr 2022; 13:1450-1461. [PMID: 35776947 PMCID: PMC9526856 DOI: 10.1093/advances/nmac075] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/31/2022] [Accepted: 06/28/2022] [Indexed: 01/28/2023] Open
Abstract
Humans often show variable responses to dietary, prebiotic, and probiotic interventions. Emerging evidence indicates that the gut microbiota is a key determinant for this population heterogeneity. Here, we provide an overview of some of the major computational and experimental tools being applied to critical questions of microbiota-mediated personalized nutrition and health. First, we discuss the latest advances in in silico modeling of the microbiota-nutrition-health axis, including the application of statistical, mechanistic, and hybrid artificial intelligence models. Second, we address high-throughput in vitro techniques for assessing interindividual heterogeneity, from ex vivo batch culturing of stool and continuous culturing in anaerobic bioreactors, to more sophisticated organ-on-a-chip models that integrate both host and microbial compartments. Third, we explore in vivo approaches for better understanding of personalized, microbiota-mediated responses to diet, prebiotics, and probiotics, from nonhuman animal models and human observational studies, to human feeding trials and crossover interventions. We highlight examples of existing, consumer-facing precision nutrition platforms that are currently leveraging the gut microbiota. Furthermore, we discuss how the integration of a broader set of the tools and techniques described in this piece can generate the data necessary to support a greater diversity of precision nutrition strategies. Finally, we present a vision of a precision nutrition and healthcare future, which leverages the gut microbiota to design effective, individual-specific interventions.
Collapse
Affiliation(s)
| | - Thomas Gurry
- Pharmaceutical Biochemistry group, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland (PSI-WS), University of Geneva/University of Lausanne, Geneva, Switzerland
| | - Johanna W Lampe
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Veerle Dam
- Sensus BV (Royal Cosun), Roosendaal, The Netherlands
| | - Amandine Everard
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Almudena Goas
- Department of Food, Nutrition, and Exercise Sciences, University of Surrey, Guildford, United Kingdom
| | - Gabriele Gross
- Medical and Scientific Affairs, Reckitt| Mead Johnson Nutrition Institute, Nijmegen, The Netherlands
| | - Michiel Kleerebezem
- Host Microbe Interactomics Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Jonathan Lane
- Health and Happiness Group, H&H Research, Cork, Ireland
| | | | - Ana Lucia Barretto Penna
- Department of Food Engineering and Technology, São Paulo State University, São José do Rio Preto, Brazil
| | - Bruno Pot
- Yakult Europe BV, Almere, The Netherlands
| | - Ana M Valdes
- Nottingham NIHR Biomedical Research Centre at the School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Gemma Walton
- Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Adrienne Weiss
- Yili Innovation Center Europe, Wageningen, The Netherlands
| | | | - Naomi V Venlet
- International Life Sciences Institute, European Branch, Brussels, Belgium
| | - Michela Miani
- International Life Sciences Institute, European Branch, Brussels, Belgium
| |
Collapse
|
29
|
Yu Y, Liu C, Liu J, Zhuang J, Sun C. Commentary: The Modulation of Chaihu Shugan Formula on Microbiota Composition in the Simulator of the Human Intestinal Microbial Ecosystem Technology Platform and Its Influence on Gut Barrier and Intestinal Immunity in Caco-2/THP1-Blue™ Cell Co-Culture Model. Front Pharmacol 2022; 13:904890. [PMID: 35721147 PMCID: PMC9198566 DOI: 10.3389/fphar.2022.904890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Yang Yu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Jingyang Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China.,Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| |
Collapse
|
30
|
El Houari A, Ecale F, Mercier A, Crapart S, Laparre J, Soulard B, Ramnath M, Berjeaud JM, Rodier MH, Crépin A. Development of an in vitro Model of Human Gut Microbiota for Screening the Reciprocal Interactions With Antibiotics, Drugs, and Xenobiotics. Front Microbiol 2022; 13:828359. [PMID: 35495704 PMCID: PMC9042397 DOI: 10.3389/fmicb.2022.828359] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/09/2022] [Indexed: 11/17/2022] Open
Abstract
Altering the gut microbiota can negatively affect human health. Efforts may be sustained to predict the intended or unintended effects of molecules not naturally produced or expected to be present within the organism on the gut microbiota. Here, culture-dependent and DNA-based approaches were combined to UHPLC-MS/MS analyses in order to investigate the reciprocal interactions between a constructed Human Gut Microbiota Model (HGMM) and molecules including antibiotics, drugs, and xenobiotics. Our HGMM was composed of strains from the five phyla commonly described in human gut microbiota and belonging to Firmicutes, Bacteroidetes, Proteobacteria, Fusobacteria, and Actinobacteria. Relevantly, the bacterial diversity was conserved in our constructed human gut model through subcultures. Uneven richness distribution was revealed and the sensitivity of the HGMM was mainly affected by antibiotic exposure rather than by drugs or xenobiotics. Interestingly, the constructed model and the individual cultured strains respond with the same sensitivity to the different molecules. UHPLC-MS/MS analyses revealed the disappearance of some native molecules in the supernatants of the HGMM as well as in those of the individual strains. These results suggest that biotransformation of molecules occurred in the presence of our gut microbiota model and the coupled approaches performed on the individual cultures may emphasize new bacterial strains active in these metabolic processes. From this study, the new HGMM appears as a simple, fast, stable, and inexpensive model for screening the reciprocal interactions between the intestinal microbiota and molecules of interest.
Collapse
Affiliation(s)
- Abdelaziz El Houari
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| | - Florine Ecale
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| | - Anne Mercier
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| | - Stéphanie Crapart
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| | | | | | | | - Jean-Marc Berjeaud
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| | - Marie-Hélène Rodier
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France.,Laboratoire de Parasitologie et Mycologie, CHU de Poitiers, Poitiers, France
| | - Alexandre Crépin
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| |
Collapse
|
31
|
Pozzo L, Alcántara C, Selma-Royo M, Garcia-Mantrana I, Bramanti E, Longo V, Collado MC, Pucci L. The impact of sourdough fermentation of spelt (Triticum dicoccum) from Garfagnana on gut microbiota composition and in vitro activity. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
32
|
Tian B, Geng Y, Xu T, Zou X, Mao R, Pi X, Wu W, Huang L, Yang K, Zeng X, Sun P. Digestive Characteristics of Hericium erinaceus Polysaccharides and Their Positive Effects on Fecal Microbiota of Male and Female Volunteers During in vitro Fermentation. Front Nutr 2022; 9:858585. [PMID: 35433782 PMCID: PMC9008368 DOI: 10.3389/fnut.2022.858585] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/15/2022] [Indexed: 12/12/2022] Open
Abstract
Hericium erinaceus polysaccharides (HEPs) have attracted widespread attention in regulating gut microbiota (GM). To investigate digestibility and fermentation of HEPs and their effects on GM composition, three polysaccharide fractions, namely, HEP-30, HEP-50, and HEP-70, were fractionally precipitated with 30%, 50%, and 70% ethanol concentrations (v/v) from hot water-soluble extracts of Hericium erinaceus, respectively. Three kinds of prepared HEPs were structurally characterized and simulated gastrointestinal digestion, and their effects on human fecal microbiota fermentations of male and female and short-chain fatty acid (SCFA) production in vitro were clarified. Under digestive conditions simulating saliva, stomach, and small intestine, HEPs were not significantly influenced and safely reached the distal intestine. After 24 h of in vitro fermentation, the content of SCFAs was significantly enhanced (p < 0.05), and the retention rates of total and reducing sugars and pH value were significantly decreased (p < 0.05). Thus, HEPs could be utilized by GM, especially HEP-50, and enhanced the relative abundance of SCFA-producing bacteria, e.g., Bifidobacterium, Faecalibacterium, Blautia, Butyricicoccus, and Lactobacillus. Furthermore, HEPs reduced the relative abundances of opportunistic pathogenic bacteria, e.g., Escherichia-Shigella, Klebsiella, and Enterobacter. This study suggests that gradual ethanol precipitation is available for the preparation of polysaccharides from Hericium erinaceus, and the extracted polysaccharide could be developed as functional foods with great development value.
Collapse
Affiliation(s)
- Baoming Tian
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
| | - Yan Geng
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
| | - Tianrui Xu
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
| | - Xianguo Zou
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
| | - Rongliang Mao
- Changshan Haofeng Agricultural Development Co., Ltd., Quzhou, China
| | - Xionge Pi
- Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Weicheng Wu
- Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Liangshui Huang
- Research Institute of Changshan Tianle Edible Fungus, Quzhou, China
| | - Kai Yang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
| | - Xiaoxiong Zeng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Peilong Sun
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
33
|
Vera C, Guerrero C, Illanes A. Trends in lactose-derived bioactives: synthesis and purification. SYSTEMS MICROBIOLOGY AND BIOMANUFACTURING 2022; 2:393-412. [PMID: 38624767 PMCID: PMC8776390 DOI: 10.1007/s43393-021-00068-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 12/11/2022]
Abstract
Lactose obtained from cheese whey is a low value commodity despite its great potential as raw material for the production of bioactive compounds. Among them, prebiotics stand out as valuable ingredients to be added to food matrices to build up functional foods, which currently represent the most active sector within the food industry. Functional foods market has been growing steadily in the recent decades along with the increasing awareness of the World population about healthy nutrition, and this is having a strong impact on lactose-derived bioactives. Most of them are produced by enzyme biocatalysis because of molecular precision and environmental sustainability considerations. The current status and outlook of the production of lactose-derived bioactive compounds is presented with special emphasis on downstream operations which are critical because of the rather modest lactose conversion and product yields that are attainable. Even though some of these products have already an established market, there are still several challenges referring to the need of developing better catalysts and more cost-effective downstream operations for delivering high quality products at affordable prices. This technological push is expected to broaden the spectrum of lactose-derived bioactive compounds to be produced at industrial scale in the near future. Graphical abstract
Collapse
Affiliation(s)
- Carlos Vera
- Department of Biology, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, (USACH), Santiago, Chile
| | - Cecilia Guerrero
- School of Biochemical Engineering, Pontificia Universidad Católica de Valparaíso (PUCV), Valparaiso, Chile
| | - Andrés Illanes
- School of Biochemical Engineering, Pontificia Universidad Católica de Valparaíso (PUCV), Valparaiso, Chile
| |
Collapse
|
34
|
Asare PT, Greppi A, Pennacchia A, Brenig K, Geirnaert A, Schwab C, Stephan R, Lacroix C. In vitro Modeling of Chicken Cecal Microbiota Ecology and Metabolism Using the PolyFermS Platform. Front Microbiol 2022; 12:780092. [PMID: 34987487 PMCID: PMC8721126 DOI: 10.3389/fmicb.2021.780092] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/15/2021] [Indexed: 12/30/2022] Open
Abstract
Continuous in vitro fermentation models provide a useful tool for a fast, reproducible, and direct assessment of treatment-related changes in microbiota metabolism and composition independent of the host. In this study, we used the PolyFermS model to mimic the conditions of the chicken cecum and evaluated three nutritive media for in vitro modeling of the chicken cecal microbiota ecology and metabolism. We observed that our model inoculated with immobilized cecal microbiota and fed with a modified Viande Levure medium (mVL-3) reached a high bacterial cell density of up to approximately 10.5 log cells per mL and stable microbiota composition, akin to the host, during 82 days of continuous operation. Relevant bacterial functional groups containing primary fibrolytic (Bacteroides, Bifidobacteriaceae, Ruminococcaceae), glycolytic (Enterococcus), mucolytic (Bacteroides), proteolytic (Bacteroides), and secondary acetate-utilizing butyrate-producing and propionate-producing (Lachnospiraceae) taxa were preserved in vitro. Besides, conserved metabolic and functional Kyoto Encyclopedia of Genes and Genomes pathways were observed between in vitro microbiota and cecal inoculum microbiota as predicted by functional metagenomics analysis. Furthermore, we demonstrated that the continuous inoculation provided by the inoculum reactor generated reproducible metabolic profiles in second-stage reactors comparable to the chicken cecum, allowing for the simultaneous investigation and direct comparison of different treatments with a control. In conclusion, we showed that PolyFermS is a suitable model for mimicking chicken cecal microbiota fermentation allowing ethical and ex vivo screening of environmental factors, such as dietary additives, on chicken cecal fermentation. We report here for the first time a fermentation medium (mVL-3) that closely mimics the substrate conditions in the chicken cecum and supports the growth and metabolic activity of the cecal bacterial akin to the host. Our PolyFermS chicken cecum model is a useful tool to study microbiota functionality and structure ex vivo.
Collapse
Affiliation(s)
- Paul Tetteh Asare
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zürich, Zurich, Switzerland
| | - Anna Greppi
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zürich, Zurich, Switzerland
| | - Alessia Pennacchia
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zürich, Zurich, Switzerland
| | - Katharina Brenig
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zürich, Zurich, Switzerland
| | - Annelies Geirnaert
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zürich, Zurich, Switzerland
| | - Clarissa Schwab
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zürich, Zurich, Switzerland
| | - Roger Stephan
- Institute for Food Hygiene and Safety, University of Zurich, Zurich, Switzerland
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zürich, Zurich, Switzerland
| |
Collapse
|
35
|
Cao Z, Guo Y, Liu Z, Zhang H, Zhou H, Shang H. Ultrasonic enzyme-assisted extraction of comfrey (Symphytum officinale L.) polysaccharides and their digestion and fermentation behaviors in vitro. Process Biochem 2022. [DOI: 10.1016/j.procbio.2021.11.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
36
|
Reboleira J, Félix R, Félix C, de Melo MMR, Silva CM, Saraiva JA, Bandarra NM, Teixeira B, Mendes R, Paulo MC, Coutinho J, Lemos MFL. Evaluating the Potential of the Defatted By-Product of Aurantiochytrium sp. Industrial Cultivation as a Functional Food. Foods 2021; 10:foods10123058. [PMID: 34945609 PMCID: PMC8701938 DOI: 10.3390/foods10123058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/01/2021] [Accepted: 12/04/2021] [Indexed: 11/16/2022] Open
Abstract
While Aurantiochytrium sp. is an increasingly popular source of polyunsaturated fatty acids (PUFAs), its extraction generates high amounts of waste, including the spent, defatted residue. The composition and bioactivities of this by-product could prove to be a major part of the sustainable valorisation of this organism within the framework of a circular economy. In this study, the defatted biomass of commercial Aurantiochytrium sp. was nutritionally characterised, and its amino acid profile was detailed. Additionally, the antioxidant and prebiotic potentials of an enzymatically digested sample of defatted Aurantiochytrium sp. were evaluated under a set of miniaturised in vitro assays. The nutritional profile of the spent Aurantiochytrium biomass revealed a protein and dietary-fibre rich product, with values reaching 26.7% and 31.0% for each, respectively. It also held high concentrations of glutamic and aspartic acid, as well as a favourable lysine/arginine ratio of 3.73. The digested samples demonstrated significant Weissela cibaria and Bifidobacterium bifidum growth-enhancing potential. Residual ferric reducing antioxidant power (FRAP) activity was likely attributed to antioxidant amino acids or peptides. The study demonstrated that some of the nutritional and functional potential that reside in the defatted Aurantiochytrium sp. waste encourages additional studies and the development of food supplements employing this resource's by-products under a biorefinery framework.
Collapse
Affiliation(s)
- João Reboleira
- MARE—Marine and Environmental Sciences Centre, ESTM, Politécnico de Leiria, 2520-641 Peniche, Portugal; (J.R.); (R.F.); (C.F.)
| | - Rafael Félix
- MARE—Marine and Environmental Sciences Centre, ESTM, Politécnico de Leiria, 2520-641 Peniche, Portugal; (J.R.); (R.F.); (C.F.)
| | - Carina Félix
- MARE—Marine and Environmental Sciences Centre, ESTM, Politécnico de Leiria, 2520-641 Peniche, Portugal; (J.R.); (R.F.); (C.F.)
| | - Marcelo M. R. de Melo
- CICECO—Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (M.M.R.d.M.); (C.M.S.)
| | - Carlos M. Silva
- CICECO—Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (M.M.R.d.M.); (C.M.S.)
| | - Jorge A. Saraiva
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Narcisa M. Bandarra
- Division of Aquaculture and Upgrading, Portuguese Institute of the Sea and Atmosphere, Rua Alfredo Magalhães Ramalho, 6, 1495-006 Lisboa, Portugal; (N.M.B.); (B.T.); (R.M.)
- CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Rua dos Bragas 289, 4050-123 Porto, Portugal
| | - Bárbara Teixeira
- Division of Aquaculture and Upgrading, Portuguese Institute of the Sea and Atmosphere, Rua Alfredo Magalhães Ramalho, 6, 1495-006 Lisboa, Portugal; (N.M.B.); (B.T.); (R.M.)
- CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Rua dos Bragas 289, 4050-123 Porto, Portugal
| | - Rogério Mendes
- Division of Aquaculture and Upgrading, Portuguese Institute of the Sea and Atmosphere, Rua Alfredo Magalhães Ramalho, 6, 1495-006 Lisboa, Portugal; (N.M.B.); (B.T.); (R.M.)
- CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Rua dos Bragas 289, 4050-123 Porto, Portugal
| | - Maria C. Paulo
- Depsiextracta Tecnologias e Biológicas, Lda., Zona Industrial do Monte da Barca Rua H, Lote 62, 2100-057 Coruche, Portugal; (M.C.P.); (J.C.)
| | - Joana Coutinho
- Depsiextracta Tecnologias e Biológicas, Lda., Zona Industrial do Monte da Barca Rua H, Lote 62, 2100-057 Coruche, Portugal; (M.C.P.); (J.C.)
| | - Marco F. L. Lemos
- MARE—Marine and Environmental Sciences Centre, ESTM, Politécnico de Leiria, 2520-641 Peniche, Portugal; (J.R.); (R.F.); (C.F.)
- Correspondence:
| |
Collapse
|
37
|
Sabater C, Calvete-Torre I, Villamiel M, Moreno FJ, Margolles A, Ruiz L. Vegetable waste and by-products to feed a healthy gut microbiota: Current evidence, machine learning and computational tools to design novel microbiome-targeted foods. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
38
|
Effects of L. plantarum HY7715 on the Gut Microbial Community and Riboflavin Production in a Three-Stage Semi-Continuous Simulated Gut System. Microorganisms 2021; 9:microorganisms9122478. [PMID: 34946080 PMCID: PMC8704370 DOI: 10.3390/microorganisms9122478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/23/2021] [Accepted: 11/26/2021] [Indexed: 12/26/2022] Open
Abstract
Probiotics should be well established in the gut, passing through the digestive tract with a high degree of viability, and produce metabolites that improve the gut environment by interacting with the gut microbiome. Our previous study revealed that the Lactiplantibacillus plantarum HY7715 strain shows good bile acid resistance and a riboflavin production capacity. To confirm the interaction between HY7715 and gut microbiome, we performed a metabolite and microbiome study using a simulated gut system (SGS) that mimics the intestinal environment. Changes in the microbiome were confirmed and compared with L. plantarum NCDO1752 as the control. After 14 days, the HY7715 treatment group showed a relatively high butyrate content compared to the control group, which showed increased acetate and propionate concentrations. Moreover, the riboflavin content was higher in the HY7715 treatment group, whereas the NCDO1752 treatment group produced only small amounts of riboflavin during the treatment period and showed a tendency to decrease during the washout stage; however, the HY7715 group produced riboflavin continuously in the ascending colon during the washout period. A correlation analysis of the genus that increased as the content of riboflavin increased revealed butyrate-producing microorganisms, such as Blautia and Flavonifractor. In conclusion, treatment with L. plantarum HY7715 induced the production and maintenance of riboflavin and the enrichment of the intestinal microbiome
Collapse
|
39
|
Duque ALRF, Demarqui FM, Santoni MM, Zanelli CF, Adorno MAT, Milenkovic D, Mesa V, Sivieri K. Effect of probiotic, prebiotic, and synbiotic on the gut microbiota of autistic children using an in vitro gut microbiome model. Food Res Int 2021; 149:110657. [PMID: 34600659 DOI: 10.1016/j.foodres.2021.110657] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 12/29/2022]
Abstract
Imbalances in gut microbiota composition occur in individuals with autism spectrum disorder (ASD). The administration of probiotics, prebiotics, and synbiotics is emerging as a potential and promising strategy for regulating the gut microbiota and improving ASD-related symptoms. We first investigated the survival of the probiotics Limosilactobacillus (L.) reuteri and Bifidobacterium (B.) longum alone, mixed and combined with a galacto-oligosaccharide (GOS) under simulated gastrointestinal conditions. Next, we evaluated the impact of probiotics (L. reuteri + B. longum), prebiotic (GOS), and synbiotic (L. reuteri + B. longum + GOS) on gut microbiota composition and metabolism of children with ASD using an in vitro fermentation model (SHIME®). The combination of L. reuteri, B. longum, and GOS showed elevated gastrointestinal resistance. The probiotic, prebiotic, and synbiotic treatments resulted in a positive modulation of the gut microbiota and metabolic activity of children with ASD. More specifically, the probiotic treatment increased the relative abundance of Lactobacillus, while the prebiotic treatment increased the relative abundance of Bifidobacterium and decreased the relative abundance of Lachnoclostridium. Changes in microbial metabolism were associated with increased short-chain fatty acid concentrations and reduced ammonium levels, particularly in the prebiotic and synbiotic treatments.
Collapse
Affiliation(s)
- Ana Luiza Rocha Faria Duque
- Department of Food and Nutrition, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Fernanda Manaia Demarqui
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Mariana Marchi Santoni
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Cleslei Fernando Zanelli
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Maria Angela Tallarico Adorno
- Department of Hydraulics and Sanitation, School of Engineering of São Carlos, University of São Paulo (USP), São Carlos, Brazil
| | - Dragan Milenkovic
- Department of Internal Medicine, UC Davis School of Medicine, University of California, Davis, United States; INRAE, UNH, Université Clermont Auvergne, St Genes Champanelle, France
| | - Victoria Mesa
- Faculty of Pharmacy, Paris University, Paris, France; Food and Human Nutrition Research Group, University of Antioquia, Medellín, Colombia
| | - Katia Sivieri
- Department of Food and Nutrition, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil.
| |
Collapse
|
40
|
Yang L, Huang J, Wu X, Li L, Cai W, Zhu L, Wang S, Song H, Zhu D, Ma T, Liu H. Interactions between gut microbiota and soy hull polysaccharides regulate the air-liquid interfacial activity. Food Hydrocoll 2021. [DOI: 10.1016/j.foodhyd.2021.106704] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
41
|
Wang S, Zhang X, Li H, Ren Y, Geng Y, Lu Z, Shi J, Xu Z. Similarities and differences of oligo/poly-saccharides' impact on human fecal microbiota identified by in vitro fermentation. Appl Microbiol Biotechnol 2021; 105:7475-7486. [PMID: 34487206 DOI: 10.1007/s00253-021-11548-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 07/26/2021] [Accepted: 08/22/2021] [Indexed: 12/14/2022]
Abstract
The dietary supplementation of prebiotics is considered a promising strategy for the modulation of gut microbiota. Due to the wide variety of animal models and tremendous inter-individual variability from human investigations, the prebiotic effect of fibers is often difficult to compare between studies. Here, the effects of 11 dietary fibers on human fecal microbiota were studied using an in vitro human fecal fermentation model under well-controlled conditions. All fibers showed positive regulatory effects on short chain fatty acids (SCFAs) and several beneficial bacteria, including Parabacteroides distasonis and Bifidobacterium spp. Cultures supplemented with xylo-oligosaccharide and konjac flour showed the highest SCFAs. According to regulatory effects, fibers were divided into three groups, with 13 indicator OTUs (operational taxonomic units) identified. Fecal microbiota regulated by isomalto-oligosaccharide and chitosan-oligosaccharide were similar to fructo-oligosaccharide and inulin outputs. As a supplement to in vivo studies, our results comprehensively summarized the similarities and distinctiveness of fibers in regulating fecal microbiota structures. KEY POINTS: • Fibers were divided into three groups based on the regulatory effects in microbiota. • Thirteen indicator OTUs were identified using pairwise comparisons. • Fiber similarities and distinctive traits in regulating microbiota effect were identified.
Collapse
Affiliation(s)
- Shanshan Wang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Science, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
| | - Xiaojuan Zhang
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China. .,Jiangsu Engineering Research Center for Bioactive Products Processing Technology, 1800 Lihu Avenue, Wuxi, 214122, China.
| | - Heng Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Science, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China.,School of Biotechnology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
| | - Yilin Ren
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Science, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
| | - Yan Geng
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Science, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
| | - Zhenming Lu
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China.,Jiangsu Engineering Research Center for Bioactive Products Processing Technology, 1800 Lihu Avenue, Wuxi, 214122, China
| | - Jinsong Shi
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Science, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
| | - Zhenghong Xu
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China. .,Jiangsu Engineering Research Center for Bioactive Products Processing Technology, 1800 Lihu Avenue, Wuxi, 214122, China.
| |
Collapse
|
42
|
Sauvaitre T, Etienne-Mesmin L, Sivignon A, Mosoni P, Courtin CM, Van de Wiele T, Blanquet-Diot S. Tripartite relationship between gut microbiota, intestinal mucus and dietary fibers: towards preventive strategies against enteric infections. FEMS Microbiol Rev 2021; 45:5918835. [PMID: 33026073 DOI: 10.1093/femsre/fuaa052] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
The human gut is inhabited by a large variety of microorganims involved in many physiological processes and collectively referred as to gut microbiota. Disrupted microbiome has been associated with negative health outcomes and especially could promote the onset of enteric infections. To sustain their growth and persistence within the human digestive tract, gut microbes and enteric pathogens rely on two main polysaccharide compartments, namely dietary fibers and mucus carbohydrates. Several evidences suggest that the three-way relationship between gut microbiota, dietary fibers and mucus layer could unravel the capacity of enteric pathogens to colonise the human digestive tract and ultimately lead to infection. The review starts by shedding light on similarities and differences between dietary fibers and mucus carbohydrates structures and functions. Next, we provide an overview of the interactions of these two components with the third partner, namely, the gut microbiota, under health and disease situations. The review will then provide insights into the relevance of using dietary fibers interventions to prevent enteric infections with a focus on gut microbial imbalance and impaired-mucus integrity. Facing the numerous challenges in studying microbiota-pathogen-dietary fiber-mucus interactions, we lastly describe the characteristics and potentialities of currently available in vitro models of the human gut.
Collapse
Affiliation(s)
- Thomas Sauvaitre
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France.,Ghent University, Faculty of Bioscience Engineering, Center for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Lucie Etienne-Mesmin
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| | - Adeline Sivignon
- Université Clermont Auvergne, UMR 1071 Inserm, USC-INRAe 2018, Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte (M2iSH), Clermont-Ferrand, France
| | - Pascale Mosoni
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| | - Christophe M Courtin
- KU Leuven, Faculty of Bioscience Engineering, Laboratory of Food Chemistry and Biochemistry & Leuven Food Science and Nutrition Research Centre (LFoRCe), Leuven, Belgium
| | - Tom Van de Wiele
- Ghent University, Faculty of Bioscience Engineering, Center for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Stéphanie Blanquet-Diot
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| |
Collapse
|
43
|
Antiproliferative Effect of Colonic Fermented Phenolic Compounds from Jaboticaba ( Myrciaria trunciflora) Fruit Peel in a 3D Cell Model of Colorectal Cancer. Molecules 2021; 26:molecules26154469. [PMID: 34361622 PMCID: PMC8347777 DOI: 10.3390/molecules26154469] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/13/2021] [Accepted: 07/21/2021] [Indexed: 01/07/2023] Open
Abstract
Jaboticaba is a Brazilian native berry described as a rich source of phenolic compounds (PC) with health promoting effects. PC from jaboticaba peel powder (JPP) have low intestinal bio-accessibility and are catabolized by gut microbiota. However, the biological implication of PC-derived metabolites produced during JPP digestion remains unclear. This study aimed to evaluate the antiproliferative effects of colonic fermented JPP (FJPP) in a 3D model of colorectal cancer (CRC) composed by HT29 spheroids. JPP samples fermented with human feces during 0, 2, 8, 24 or 48 h were incubated (10,000 µg mL−1) with spheroids, and cell viability was assessed after 72 h. Chemometric analyses (cluster and principal component analyses) were used to identify the main compounds responsible for the bioactive effect. The antiproliferative effect of FJPP in the CRC 3D model was increased between 8 h and 24 h of incubation, and this effect was associated with HHDP-digalloylglucose isomer and dihydroxyphenyl-γ-valerolactone. At 48 h of fermentation, the antiproliferative effect of FJPP was negligible, indicating that the presence of urolithins did not improve the bioactivity of JPP. These findings provide relevant knowledge on the role of colonic microbiota fermentation to generate active phenolic metabolites from JPP with positive impact on CRC.
Collapse
|
44
|
Sasaki K, Mori T, Hoshi N, Sasaki D, Inoue J, Shinkura R, Kondo A. W27 IgA suppresses growth of Escherichia in an in vitro model of the human intestinal microbiota. Sci Rep 2021; 11:14627. [PMID: 34272464 PMCID: PMC8285510 DOI: 10.1038/s41598-021-94210-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 07/05/2021] [Indexed: 01/04/2023] Open
Abstract
W27 monoclonal immunoglobulin A (IgA) suppresses pathogenic Escherichia coli cell growth; however, its effect on the human intestine remains unclear. We aimed to determine how W27 IgA affects the human colonic microbiota using the in vitro microbiota model. This model was established using fecal samples collected from 12 healthy volunteers; after anaerobic cultivation, each model was found to retain the genera found in the original human fecal samples. After pre-incubating W27 IgA with the respective fecal sample under aerobic conditions, the mixture of W27 IgA (final concentration, 0.5 μg/mL) and each fecal sample was added to the in vitro microbiota model and cultured under anaerobic conditions. Next-generation sequencing of the bacterial 16S rRNA gene revealed that W27 IgA significantly decreased the relative abundance of bacteria related to the genus Escherichia in the model. Additionally, at a final concentration of 5 μg/mL, W27 IgA delayed growth in the pure culture of Escherichia coli isolated from human fecal samples. Our study thus revealed the suppressive effect of W27 IgA on the genus Escherichia at relatively low-concentrations and the usefulness of an in vitro microbiota model to evaluate the effect of IgA as a gut microbiota regulator.
Collapse
Affiliation(s)
- Kengo Sasaki
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo, 657-8501, Japan.
- BioPalette Co., Ltd., 6-3-7 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
| | - Tomoyuki Mori
- Laboratory of Immunology and Infection Control, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Namiko Hoshi
- Division of Gastroenterology, Department of Internal Medicine, Graduate School of Medicine, Kobe University, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Daisuke Sasaki
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo, 657-8501, Japan
| | - Jun Inoue
- Division of Gastroenterology, Department of Internal Medicine, Graduate School of Medicine, Kobe University, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Reiko Shinkura
- Laboratory of Immunology and Infection Control, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
- Core Research for Evolutional Science and Technology, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Akihiko Kondo
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo, 657-8501, Japan
- RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| |
Collapse
|
45
|
Fournier E, Roussel C, Dominicis A, Ley D, Peyron MA, Collado V, Mercier-Bonin M, Lacroix C, Alric M, Van de Wiele T, Chassard C, Etienne-Mesmin L, Blanquet-Diot S. In vitro models of gut digestion across childhood: current developments, challenges and future trends. Biotechnol Adv 2021; 54:107796. [PMID: 34252564 DOI: 10.1016/j.biotechadv.2021.107796] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 02/08/2023]
Abstract
The human digestion is a multi-step and multi-compartment process essential for human health, at the heart of many issues raised by academics, the medical world and industrials from the food, nutrition and pharma fields. In the first years of life, major dietary changes occur and are concomitant with an evolution of the whole child digestive tract anatomy and physiology, including colonization of gut microbiota. All these phenomena are influenced by child exposure to environmental compounds, such as drugs (especially antibiotics) and food pollutants, but also childhood infections. Due to obvious ethical, regulatory and technical limitations, in vivo approaches in animal and human are more and more restricted to favor complementary in vitro approaches. This review summarizes current knowledge on the evolution of child gut physiology from birth to 3 years old regarding physicochemical, mechanical and microbial parameters. Then, all the available in vitro models of the child digestive tract are described, ranging from the simplest static mono-compartmental systems to the most sophisticated dynamic and multi-compartmental models, and mimicking from the oral phase to the colon compartment. Lastly, we detail the main applications of child gut models in nutritional, pharmaceutical and microbiological studies and discuss the limitations and challenges facing this field of research.
Collapse
Affiliation(s)
- Elora Fournier
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, CRNH Auvergne, 63000 Clermont-Ferrand, France; Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, 31000 Toulouse, France
| | - Charlène Roussel
- Laval University, INAF Institute of Nutrition and Functional Foods, G1V 0A6 Quebec, Canada
| | - Alessandra Dominicis
- European Reference Laboratory for E. coli, Istituto Superiore di Sanità, Rome, Italy
| | - Delphine Ley
- Université Lille 2, Faculté de Médecine, Inserm U995 Nutritional Modulation of Infection and Inflammation, 59045 Lille, France
| | - Marie-Agnès Peyron
- Université Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Valérie Collado
- Université Clermont Auvergne, EA 4847, CROC, Centre de Recherche en Odontologie Clinique, 63000 Clermont-Ferrand, France
| | - Muriel Mercier-Bonin
- Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, 31000 Toulouse, France
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, 8092 Zürich, Switzerland
| | - Monique Alric
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Tom Van de Wiele
- Ghent University, Center for Microbial Ecology and Technology (CMET), Coupure Links 653, 9000 Ghent, Belgium
| | - Christophe Chassard
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, 15000 Aurillac, France
| | - Lucie Etienne-Mesmin
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Stéphanie Blanquet-Diot
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, CRNH Auvergne, 63000 Clermont-Ferrand, France.
| |
Collapse
|
46
|
Pérez-Burillo S, Molino S, Navajas-Porras B, Valverde-Moya ÁJ, Hinojosa-Nogueira D, López-Maldonado A, Pastoriza S, Rufián-Henares JÁ. An in vitro batch fermentation protocol for studying the contribution of food to gut microbiota composition and functionality. Nat Protoc 2021; 16:3186-3209. [PMID: 34089022 DOI: 10.1038/s41596-021-00537-x] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 03/18/2021] [Indexed: 02/05/2023]
Abstract
Knowledge of the effect of foods on gut microbiota composition and functionality is expanding. To isolate the effect of single foods and/or single nutrients (i.e., fiber, polyphenols), this protocol describes an in vitro batch fermentation procedure to be carried out after an in vitro gastrointestinal digestion. Therefore, this is an extension of the previous protocol described by Brodkorb et al. (2019) for studying in vitro digestion. The current protocol uses an oligotrophic fermentation medium with peptone and a high concentration of fecal inoculum from human fecal samples both to provide the microbiota and as the main source of nutrients for the bacteria. This protocol is recommended for screening work to be performed when many food samples are to be studied. It has been used successfully to study gut microbiota fermentation of different foodstuffs, giving insights into their functionality, community structure or ability to degrade particular substances, which can contribute to the development of personalized nutrition strategies. The procedure does not require a specific level of expertise. The protocol takes 4-6 h for preparation of fermentation tubes and 20 h for incubation.
Collapse
Affiliation(s)
- Sergio Pérez-Burillo
- Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de Alimentos, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Silvia Molino
- Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de Alimentos, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Beatriz Navajas-Porras
- Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de Alimentos, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Álvaro Jesús Valverde-Moya
- Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de Alimentos, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Daniel Hinojosa-Nogueira
- Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de Alimentos, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Alicia López-Maldonado
- Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de Alimentos, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Silvia Pastoriza
- Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de Alimentos, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - José Ángel Rufián-Henares
- Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de Alimentos, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain. .,Instituto de Investigación Biosanitaria ibs.GRANADA, Universidad de Granada, Granada, Spain.
| |
Collapse
|
47
|
Affiliation(s)
- Zhonglin Zhao
- College of Sciences Henan Agricultural University Zhengzhou 450002 P.R. China
| | - Wei Liu
- Zhejiang Academy of Agricultural Sciences Hangzhou 310021 P.R. China
| | - Xionge Pi
- Zhejiang Academy of Agricultural Sciences Hangzhou 310021 P.R. China
| |
Collapse
|
48
|
Roupar D, Berni P, Martins JT, Caetano AC, Teixeira JA, Nobre C. Bioengineering approaches to simulate human colon microbiome ecosystem. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.04.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
49
|
Salgaço MK, Perina NP, Tomé TM, Mosquera EMB, Lazarini T, Sartoratto A, Sivieri K. Probiotic infant cereal improves children's gut microbiota: Insights using the Simulator of Human Intestinal Microbial Ecosystem (SHIME®). Food Res Int 2021; 143:110292. [PMID: 33992391 DOI: 10.1016/j.foodres.2021.110292] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 02/27/2021] [Accepted: 02/27/2021] [Indexed: 02/08/2023]
Abstract
Infant́s gut microbiota can be modulated by many factors, including mode of delivery, feeding regime, maternal diet/weight and probiotic and prebiotic consumption. The gut microbiota in dysbiosis has been associated with innumerous diseases. In this sense, early childhood intestinal microbiome modulation can be a strategy for disease prevention. This study had the purpose to evaluate the effect of an infant cereal with probiotic (Bifidobacterium animalis ssp. lactis BB-12®) on infant́s intestinal microbiota using SHIME®, which simulates human gastrointestinal conditions. The ascending colon was inoculated with fecal microbiota from three children (2-3 years old). NH4+, short chain fatty acids (SCFASs) and microbiota composition were determined by selective ion electrode, GC/MS and 16S sequencing, respectively. After treatment, butyric acid production increased (p < 0.05) 52% and a decrease in NH4+ production was observed (p < 0.01). The treatment stimulated an increase (p < 0.01) of Lactobacillaceae families, more precisely L. gasseri and L. kefiri. L. gasseri has been associated with the prevention of allergic rhinitis in children and L. kefiri in the prevention of obesity. Thus, infant cereal with BB-12® is able to stimulate the growth of L. gasseri and L. kefiri in a beneficial way, reducing NH4+ and increasing the production of SCFAs, especially butyric acid, in SHIME®.
Collapse
Affiliation(s)
- Mateus Kawata Salgaço
- Department of Food and Nutrition, School of Pharmaceutical Science, São Paulo State University-UNESP, Araraquara, São Paulo, Brazil
| | - Natália Partis Perina
- Medical, Scientific and Regulatory Affairs - Nestlé Nutrition/Nestlé Brazil Ltda, São Paulo, Brazil
| | - Thaís Moreno Tomé
- Medical, Scientific and Regulatory Affairs - Nestlé Nutrition/Nestlé Brazil Ltda, São Paulo, Brazil
| | | | - Tamara Lazarini
- Medical, Scientific and Regulatory Affairs - Nestlé Nutrition/Nestlé Brazil Ltda, São Paulo, Brazil
| | | | - Katia Sivieri
- Department of Food and Nutrition, School of Pharmaceutical Science, São Paulo State University-UNESP, Araraquara, São Paulo, Brazil.
| |
Collapse
|
50
|
Effects of Polyphenols in Tea (Camellia sinensis sp.) on the Modulation of Gut Microbiota in Human Trials and Animal Studies. GASTROENTEROLOGY INSIGHTS 2021. [DOI: 10.3390/gastroent12020018] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A diet high in polyphenols is associated with a diversified gut microbiome. Tea is the second most consumed beverage in the world, after water. The health benefits of tea might be attributed to the presence of polyphenol compounds such as flavonoids (e.g., catechins and epicatechins), theaflavins, and tannins. Although many studies have been conducted on tea, little is known of its effects on the trillions of gut microbiota. Hence, this review aimed to systematically study the effect of tea polyphenols on the stimulation or suppression of gut microbiota in humans and animals. It was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) protocol. Articles were retrieved from PubMed and Scopus databases, and data were extracted from 6 human trials and 15 animal studies. Overall, large variations were observed in terms of microbiota composition between humans and animals. A more consistent pattern of diversified microbiota was observed in animal studies. Tea alleviated the gut microbiota imbalance caused by high-fat diet-induced obesity, diabetes, and ultraviolet-induced damage. The overall changes in microbiota composition measured by beta diversity analysis showed that tea had shifted the microbiota from the pattern seen in animals that received tea-free intervention. In humans, a prebiotic-like effect was observed toward the gut microbiota, but these results appeared in lower-quality studies. The beta diversity in human microbiota remains intact despite tea intervention; supplementation with different teas affects different types of bacterial taxa in the gut. These studies suggest that tea polyphenols may have a prebiotic effect in disease-induced animals and in a limited number of human interventions. Further intervention is needed to identify the mechanisms of action underlying the effects of tea on gut microbiota.
Collapse
|