1
|
Pessotti RDC, Guerville M, Agostinho LL, Bogsan CSB, Salgaço MK, Ligneul A, Freitas MND, Guimarães CRW, Sivieri K. Bugs got milk? Exploring the potential of lactose as a prebiotic ingredient for the human gut microbiota of lactose-tolerant individuals. Nutr Res 2025; 136:64-80. [PMID: 40154186 DOI: 10.1016/j.nutres.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 04/01/2025]
Abstract
Milk consumption is important to help meet daily nutrient requirements. However, lactose-present in dairy products-has been associated with digestive discomfort in individuals who are lactose intolerant or have inadequate lactase activity. Yet, a new perspective on this dietary component has emerged: its potential as a prebiotic for the lactose-tolerant population. We hypothesized that ingestion of lactose may improve the microbial community structure and metabolism of the gut microbiota from healthy adults. First, we assessed the acute impact of lactose ingestion on the gut microbiota of adults using a short-duration in vitro batch colonic model. Subsequently, we employed a long-duration in vitro dynamic multivessel colonic model to evaluate the effects of lactose chronic ingestion. In both cases, a mixture of lactose/galactose/glucose was administered in a defined proportion to mimic lactose metabolism and galactose/glucose absorption in lactose-tolerant adults. The hypothesis was confirmed, as a modulatory prebiotic effect was revealed on the microbial community structure and metabolism of the microbiota upon treatments simulating the ingestion of three doses of lactose, equivalent to half a glass, one glass, and two glasses of cow's milk. The long-duration model confirmed this potential, increasing the relative abundance of the beneficial genera Lactobacillus, Akkermansia, and Faecalibacterium, while the usually detrimental genus Clostridium decreased. Additionally, the health-promoting microbial metabolites acetate, propionate, and lactate were increased. Therefore, lactose ingestion could positively modulate the gut microbiota in healthy lactose-tolerant adults, thereby promoting gut health and shedding light on the dietary benefits of consuming milk.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Katia Sivieri
- Nintx-Next Innovative Therapeutics, São Paulo, Brazil; Department of Food and Nutrition, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil; Department of Biotechnology, University of Araraquara (UNIARA), Araraquara, Brazil
| |
Collapse
|
2
|
Govaert M, Duysburgh C, Kesler B, Marzorati M. Effects of NatureKnit™, a Blend of Fruit and Vegetable Fibers Rich in Naturally Occurring Bound Polyphenols, on the Metabolic Activity and Community Composition of the Human Gut Microbiome Using the M-SHIME ® Gastrointestinal Model. Microorganisms 2025; 13:613. [PMID: 40142506 PMCID: PMC11944907 DOI: 10.3390/microorganisms13030613] [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: 02/05/2025] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
This study evaluated the impact of a proprietary blend of fruit and vegetable fibers rich in naturally occurring bound polyphenols (commercially marketed as NatureKnitTM), compared to purified fibers (inulin and psyllium), on the human gut microbiome using the validated M-SHIME® gastrointestinal model. A short-term single-stage colonic M-SHIME® experiment (with fecal inoculum from three healthy human donors) was used to evaluate the test products compared to a negative control. Samples were assessed for pH, gas pressure, short-chain fatty acid (SCFA) production, lactate, and ammonium from 0 h to 48 h. Microbial community composition was assessed at 0 h (negative control only), 24 h, and 48 h (lumen) or 48 h (mucosal). All test products were fermented well in the colon as demonstrated by decreases in pH and increases in gas pressure over time; these changes occurred faster with the purified fibers, whereas NatureKnit™ demonstrated slow, steady changes, potentially indicating a gentler fermentation process. SCFA production significantly increased over the course of the 48 h experiment with all test products versus negative control. SCFA production was significantly greater with NatureKnit™ versus the purified fibers. Shifts in the microbial community composition were observed with all test products versus negative control. At the conclusion of the 48 h experiment, the absolute bacterial abundance and the richness of observed bacterial taxa in the lumen compartment was significantly greater with NatureKnit™ compared with inulin, psyllium, and negative control. Overall, NatureKnit™ demonstrated greater or similar prebiotic effects on study measures compared with established prebiotic fibers.
Collapse
Affiliation(s)
- Marlies Govaert
- ProDigest, Technologiepark 82, 9052 Zwijnaarde, Belgium; (M.G.); (C.D.)
| | - Cindy Duysburgh
- ProDigest, Technologiepark 82, 9052 Zwijnaarde, Belgium; (M.G.); (C.D.)
| | - Brendan Kesler
- VDF FutureCeuticals, Inc., 2692 N. State Rt. 1-17, Momence, IL 60954, USA;
| | - Massimo Marzorati
- ProDigest, Technologiepark 82, 9052 Zwijnaarde, Belgium; (M.G.); (C.D.)
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Gent, Belgium
| |
Collapse
|
3
|
Yang D, Luo F, Wu M, Zhang Z, Luo J, Zhao Z, Guo L. Establishment of a Low-Cost and Efficient In Vitro Model for Cultivating Intestinal Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:2451-2460. [PMID: 39829030 DOI: 10.1021/acs.jafc.4c07754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The Simulator of Human Intestinal Microbial Ecosystem (SHIME) has hindered widespread adoption due to its high cost. This study founded biomimetic multilink fermentation equipment (BMLFE), priced at half or even lower than SHIME. It was improved based on multilink fermentation equipment (MLFE) by modifying materials, peristaltic pumps, fermentation time, and dietary habits while calculating transfer time and volumes and conducted anaerobic fermentation for 15 days followed by monitoring changes in intestinal microbial composition and short-chain fatty acids (SCFAs). We observed that the intestinal microbiota achieved a stable state after the ninth day and retained the predominant bacterial species in the fecal inoculum. The Bacillota/Bacteroidota values of the descending colon (DC) were similar to those in the fecal samples. However, the stability of SCFAs was relatively delayed and reached stability only after the 11th day. Meanwhile, the concentration ratio of acetic acid, propionate, and butyric acid metabolized by transverse colon (TC) and DC on the 11-15th days was close to that in fecal inoculations. Therefore, BMLFE can be used to simulate the human gastrointestinal environment in vitro studies. It is expected to be employed in clinical FMT and may even contribute to establishing stable enterotypes through dietary intervention.
Collapse
Affiliation(s)
- Dayong Yang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Fudi Luo
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Mingdian Wu
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Zeyu Zhang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Junjie Luo
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Zuguo Zhao
- Teaching and Research Office of Microbiology and Immunology, School of Basic Medical Sciences, Guangdong Medical University, Dongguan 523808, China
| | - Lianxian Guo
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
4
|
Yang W, Pan Z, Zhang J, Wang L, Lai J, Fan K, Zhu J, Liu Q, Dai Y, Zhou J, Wu S, Gao Z, Yu S. Administration Strategy-Dependent Mechanisms and Effects of Human Adipose Tissue Stem Cell Extracellular Vesicles in Mouse Allergic Rhinitis Treatment. Cell Transplant 2025; 34:9636897251325673. [PMID: 40179013 PMCID: PMC11970061 DOI: 10.1177/09636897251325673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 04/05/2025] Open
Abstract
We previously found that intravenous injection of extracellular vesicles (EVs) from human adipose tissue-derived stem cells (hADSC) could ameliorate allergic rhinitis (AR) in mice through immunomodulatory effects. In clinical trials, nasal delivery has been an attractive treatment for AR. We sought to determine whether there are differences in the therapeutic effects between caudal injection and their combination. We treated AR mice with ADSC-EVs via caudal vein, nasal cavity, or both. After treatment, the mice were re-sensitized and the indices of behavior, nasal mucosa morphology, and cytokine secretion of the mice under different modes of administration were calculated. The resultes show that tail vein, nasal, and combined administration could effectively relieve the inflammatory infiltration of the nasal mucosa of mice, reduce the secretion of IgE, IL-4, and other inflammatory factors, and alleviate the Th1/Th2 imbalance. Injection and nasal delivery, as well as their combination, effectively alleviated the symptoms of rhinitis in mice. Nasal administration has a better therapeutic effect when the inflammatory response is mild. It could be speculated that ADSC-EVs have excellent properties in the treatment of AR, and modes of administration can be selected for different stages of treatment in clinical therapy.
Collapse
Affiliation(s)
- Wenhan Yang
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Anesthesiology, Shanghai Gongli Hospital, Naval Military Medical University, Shanghai, China
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
| | - Zhiyu Pan
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiacheng Zhang
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lian Wang
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ju Lai
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Anesthesiology, Shanghai Gongli Hospital, Naval Military Medical University, Shanghai, China
| | - Kai Fan
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jingjing Zhu
- Department of Otorhinolaryngology, Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qian Liu
- Department of Urology, Tianjin First Central Hospital, Tianjin, China
| | - Yalei Dai
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jieyu Zhou
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shuhui Wu
- Department of Otorhinolaryngology, Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhengliang Gao
- Department of Anesthesiology, Shanghai Gongli Hospital, Naval Military Medical University, Shanghai, China
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
- China-Japan Friendship Medical Research Institute, Shanghai University, Shanghai, China
| | - Shaoqing Yu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Anesthesiology, Shanghai Gongli Hospital, Naval Military Medical University, Shanghai, China
- Department of Allergy, School of Medicine, Tongji Hospital, Tongji University, Shanghai, China
| |
Collapse
|
5
|
Zhu W, Zhang X, Wang D, Yao Q, Ma GL, Fan X. Simulator of the Human Intestinal Microbial Ecosystem (SHIME ®): Current Developments, Applications, and Future Prospects. Pharmaceuticals (Basel) 2024; 17:1639. [PMID: 39770481 PMCID: PMC11677124 DOI: 10.3390/ph17121639] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/28/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
The human gastrointestinal microbiota plays a vital role in maintaining host health and preventing diseases, prompting the creation of simulators to replicate this intricate system. The Simulator of the Human Intestinal Microbial Ecosystem (SHIME®), a multicompartment dynamic simulator, has emerged as a pivotal in vitro model for studying the interactions and interferences within the human gut microbiota. The continuous and real-time monitoring hallmarks, along with the programmatically flexible setup, bestow SHIME® with the ability to mimic the entire human intestinal ecosystem with high dynamics and stability, allowing the evaluation of various treatments on the bowel microbiota in a controlled environment. This review outlines recent developments in SHIME® systems, including the M-SHIME®, Twin-SHIME®, Triple-SHIME®, and Toddle SHIME® models, highlighting their applications in the fields of food and nutritional science, drug development, gut health research, and traditional Chinese medicine. Additionally, the prospect of SHIME® integrating with other advanced technologies is also discussed. The findings underscore the versatility of SHIME® technology, demonstrating its significant contributions to current gut ecosystem research and its potential for future innovation in microbiome-related fields.
Collapse
Affiliation(s)
- Wei Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (W.Z.); (G.-L.M.)
| | - Xiaoyong Zhang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310000, China;
| | - Dong Wang
- Department of Orthopaedics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, China;
| | - Qinghua Yao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310005, China;
| | - Guang-Lei Ma
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (W.Z.); (G.-L.M.)
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314102, China
| | - Xiaohui Fan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (W.Z.); (G.-L.M.)
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314102, China
- The Joint-Laboratory of Clinical Multi-Omics Research Between Zhejiang University and Ningbo Municipal Hospital of TCM, Ningbo Municipal Hospital of TCM, Ningbo 315010, China
| |
Collapse
|
6
|
Li J, Shen N, He W, Pan Y, Wu J, Zhao R, Mo X, Li Y. Gut microbiome impact on childhood allergic rhinitis and house dust mite IgE responses. Pediatr Res 2024:10.1038/s41390-024-03645-y. [PMID: 39433961 DOI: 10.1038/s41390-024-03645-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND The correlation between the gut microbiota and airway inflammation in childhood allergic rhinitis (AR), particularly concerning allergen exposure, remains insufficiently explored. This study aimed to link gut microbiota changes with house dust mite (HDM)-specific IgE responses in pediatric AR. METHODS Using metagenomic shotgun sequencing, we compared the fecal microbiota of 60 children with HDM-AR to 48 healthy controls (HC), analyzing the link to IgE reactions. We examined the effects of oral Escherichia (E.) fergusonii treatment in mice sensitized with ovalbumin and HDM on allergic symptoms, mucosal cell infiltration, Th1/Th2/Tregs balance in the spleen, serum cytokine levels, and E. fergusonii presence in feces. RESULTS Children with HDM-AR have a less diverse gut microbiome and lower levels of E. fergusonii compared to controls, with a negative correlation between E. fergusonii abundance and HDM-specific IgE levels. In mice sensitized with OVA and HDM, oral administration of E. fergusonii improved allergic symptoms, reduced nasal eosinophils/mast cells infiltration and adjusted Th cell populations towards a non-allergic profile in splenic lymphocytes with exception of IFN-γ change in serum. CONCLUSION These findings underline the potential of targeting gut microbiota, particularly E. fergusonii, in managing childhood HDM-AR, suggesting a promising approach for future interventions. IMPACT The composition and distribution of gut microbiota in children with HDM-AR are significant changed. The abundance of Escherichia genus is decreased in HDM-AR children. HDM-specific IgE levels are strongly negatively associated with E. fergusonii abundance. Oral administration of E. fergusonii effectively suppresses allergic responses in murine model. These findings offer novel insights into the diagnosis and treatment of HDM-AR, which suggested that E. fergusonii holds promise as a potential therapeutic avenue for managing HDM-AR.
Collapse
Affiliation(s)
- Junyang Li
- Department of Otolaryngology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Nan Shen
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Infectious Diseases, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenjun He
- Department of Otolaryngology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Pan
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Wu
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruike Zhao
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xi Mo
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Youjin Li
- Department of Otolaryngology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Department of Otolaryngology, Hainan Branch, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China.
| |
Collapse
|
7
|
Nieto JA, Rosés C, García-Ibáñez P, Pérez B, Viadel B, Romo-Hualde A, Milagro FI, Barceló A, Carvajal M, Gallego E, Agudelo A. Fiber from elicited butternut pumpkin (Cucurbita moschata D. cv. Ariel) modulates the human intestinal microbiota dysbiosis. Int J Biol Macromol 2024; 269:132130. [PMID: 38723828 DOI: 10.1016/j.ijbiomac.2024.132130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 04/30/2024] [Accepted: 05/05/2024] [Indexed: 05/12/2024]
Abstract
Elicited pumpkin was evaluated as a potential daily consumption product able to modulate the gut microbiota. An in vitro dynamic colonic fermentation performance with microbiota from obese volunteers was used. Prebiotic effects were observed after the pumpkin treatment. Bifidobacterium abundance was maintained during the treatment period whereas Lactobacillus increased in the transversal and descending colon. Conversely, Enterobacteriaceae and Clostridium groups were more stable, although scarce decreasing trends were observed for same species. Increments of Lactobacillus acidophilus and Limosilactobacillus fermentum (old Lactobacillus fermentum) were observed in the whole colonic tract after the treatment period. However, modulatory effects were mainly observed in the transversal and descending colon. Diverse bacteria species were increased, such as Akkermansia muciniphila, Bacteroides dorei, Cloacibacillus porcorum, Clostridium lactatifermentans, Ruminococcus albus, Ruminococcus lactaris, Coprococcus catus, Alistipes shahii or Bacteroides vulgatus. The prebiotic effect of the elicited pumpkin was provided by the fiber of the pumpkin, suggesting a release of pectin molecules in the transversal and distal colonic tract through low cellulosic fiber degradation, explaining the increases in the total propionic and butyric acid in these colonic sections. Also, a possible modulatory role of carotenoids from the sample was suggested since carotenes were found in the descending colon. Hence, the results of this research highlighted pumpkin as a natural product able to modulate the microbiota towards a healthier profile.
Collapse
Affiliation(s)
- Juan Antonio Nieto
- Centro Tecnológico Ainia, Parque Tecnológico de Valencia, E46980 Paterna, Spain; Bioactivity and Nutritional Immunology Group (BIOINUT), Faculty of Health Science, Universidad Internacional de Valencia (VIU), Calle Pintor Sorolla 21, E46002, Valencia, Spain.
| | - Carles Rosés
- Servei de Genòmica, Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallés, Spain
| | - Paula García-Ibáñez
- Group of Aquaporins, Plant Nutrition Department, Centro de Edafologia y Biologia Aplicada del Segura, CEBAS-CSIC, Campus Universitario de Espinardo, E-30100 Murcia, Spain
| | - Beatriz Pérez
- Centro Tecnológico Ainia, Parque Tecnológico de Valencia, E46980 Paterna, Spain
| | - Blanca Viadel
- Centro Tecnológico Ainia, Parque Tecnológico de Valencia, E46980 Paterna, Spain
| | - Ana Romo-Hualde
- Department of Nutrition, Food Science and Physiology, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain; Centre for Nutrition Research, Department of Nutrition, Food Sciences and Physiology, University of Navarra, 31008 Pamplona, Spain
| | - Fermín I Milagro
- Department of Nutrition, Food Science and Physiology, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain; Centre for Nutrition Research, Department of Nutrition, Food Sciences and Physiology, University of Navarra, 31008 Pamplona, Spain; Navarra Institute for Health Research (IdISNA), 31008 Pamplona, Spain; Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Anna Barceló
- Servei de Genòmica, Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallés, Spain
| | - Micaela Carvajal
- Group of Aquaporins, Plant Nutrition Department, Centro de Edafologia y Biologia Aplicada del Segura, CEBAS-CSIC, Campus Universitario de Espinardo, E-30100 Murcia, Spain
| | - Elisa Gallego
- Centro Tecnológico Ainia, Parque Tecnológico de Valencia, E46980 Paterna, Spain
| | - Agatha Agudelo
- Sakata Seed Ibérica S.L.U., Pl/ Poeta Vicente Gaos, 6 Bajo. 46021, Valencia, Spain
| |
Collapse
|
8
|
Asensio-Grau A, Heredia A, García-Hernández J, Cabrera-Rubio R, Masip E, Ribes-Koninckx C, Collado MC, Andrés A, Calvo-Lerma J. Effect of beta-glucan supplementation on cystic fibrosis colonic microbiota: an in vitro study. Pediatr Res 2024; 95:1519-1527. [PMID: 38092964 DOI: 10.1038/s41390-023-02944-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/06/2023] [Accepted: 11/18/2023] [Indexed: 05/26/2024]
Abstract
BACKGROUND Children with cystic fibrosis (CF) present with gut dysbiosis, and current evidence impedes robust recommendations on the use of prebiotics. This study aimed at establishing the prebiotic potential of a commercial beta-glucan on the in vitro colonic microbiota of a child with CF compared to a healthy counterpart (H). METHODS A dynamic simulator of colonic fermentation (twin-SHIME® model) was set up including the simulation of the proximal (PC) and distal colon (DC) of the CF and the H subjects by colonizing the bioreactors with faecal microbiota. During two weeks the system was supplied with the beta-glucan. At baseline, during treatment and post-treatment, microbiota composition was profiled by 16 S rRNA and short-chain fatty acids (SCFA) production was determined by GS-MS. RESULTS At baseline, Faecalibacterium, was higher in CF' DC than in the H, along higher Acidaminococcus and less Megasphaera and Sutterella. Beta-glucan supplementation induced increased microbiota richness and diversity in both subjects during the treatment. At genus level, Pseudomonas and Veillonella decreased, while Akkermansia and Faecalibacterium increased significantly in CF. CONCLUSION The supplementation with beta-glucan suggests positive results on CF colonic microbiota in the in vitro context, encouraging further research in the in vivo setting. IMPACT Current evidence supports assessing the effect of prebiotics on modifying cystic fibrosis microbiota. The effect of beta-glucan supplementation was evaluated in a controlled dynamic in vitro colonic ecosystem. Beta-glucan supplement improved diversity in cystic fibrosis colonic microbiota. The treatment showed increased abundance of Faecalibacterium and Akkermansia in cystic fibrosis. New evidence supports the use of prebiotics in future clinical studies.
Collapse
Affiliation(s)
- Andrea Asensio-Grau
- Instituto de Ingeniería de Alimentos para el Desarrollo. Universitat Politècnica de València, València, Spain.
- Unidad Mixta de Investigación (NutriCuraPDig), Valencia, Spain.
| | - Ana Heredia
- Instituto de Ingeniería de Alimentos para el Desarrollo. Universitat Politècnica de València, València, Spain
- Unidad Mixta de Investigación (NutriCuraPDig), Valencia, Spain
| | - Jorge García-Hernández
- Unidad Mixta de Investigación (NutriCuraPDig), Valencia, Spain
- Centro Avanzado de Microbiología de Alimentos (CAMA), Universitat Politècnica de València, Valencia, Spain
| | - Raúl Cabrera-Rubio
- Institute of Agrochemistry and Food Technology-Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Etna Masip
- Unidad Mixta de Investigación (NutriCuraPDig), Valencia, Spain
- Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Carmen Ribes-Koninckx
- Unidad Mixta de Investigación (NutriCuraPDig), Valencia, Spain
- Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Ana Andrés
- Instituto de Ingeniería de Alimentos para el Desarrollo. Universitat Politècnica de València, València, Spain
- Unidad Mixta de Investigación (NutriCuraPDig), Valencia, Spain
| | - Joaquim Calvo-Lerma
- Unidad Mixta de Investigación (NutriCuraPDig), Valencia, Spain
- Institute of Agrochemistry and Food Technology-Spanish National Research Council (IATA-CSIC), Valencia, Spain
| |
Collapse
|
9
|
Gomes de Oliveira LI, Clementino JR, Salgaço MK, de Oliveira SPA, Dos Santos Lima M, Mesa V, de Souza EL, Vinderola CG, Magnani M, Sivieri K. Revealing the beneficial effects of a dairy infant formula on the gut microbiota of early childhood children with autistic spectrum disorder using static and SHIME® fermentation models. Food Funct 2023; 14:8964-8974. [PMID: 37724612 DOI: 10.1039/d3fo01156a] [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: 09/21/2023]
Abstract
This study evaluated the impact of the Milnutri Profutura® (MNP) dairy infant formula on the gut microbiota of early childhood children (three to five years) with Autistic Spectrum Disorder (ASD) using static fermentation (time zero, 24, and 48 h) and the Simulator of the Human Intestinal Microbiol Ecosystem (SHIME®) (time zero, 72 h, and 7 days). The relative abundance of selected intestinal bacterial groups, pH values, organic acids, and sugars were verified at time zero, 24, and 48 h using flow cytometry and measurements. In addition, the diversity and changes in the gut microbiota, and the amounts of acetic, butyric, and propionic acids and ammonium ions (NH4+) in fermentation using the SHIME® were measured at time zero, 72 h, and 7 days. MNP increased Lactobacillus/Enterococcus and Bifidobacterium populations and decreased Bacteroides/Prevotella, Clostridium histolyticum and Eubacterium rectale/Clostridium coccoides populations (p < 0.05) at 24 and 48 h of static fermentation, showing a positive prebiotic activity score (65.18 ± 0.07). The pH, fructose and glucose decreased, while lactic, butyric, and propionic acids increased (p < 0.05) at 48 h of static fermentation. MNP increased (p < 0.05) the Firmicutes phylum during the fermentation in SHIME®. MNP decreased the diversity at 72 h of fermentation, mostly by the increase (p < 0.05) in the Lactobacillus genus. Microbial groups considered harmful such as Lachnospiraceae, Negativicoccus, and Lachnoclostridium were inhibited after administration with MNP. Propionic and butyric acids increased at 72 h and NH4+ decreased (p < 0.05) at the end of fermentation with MNP. The results indicate MNP as an infant formula which may benefit the gut microbiota of children with ASD.
Collapse
Affiliation(s)
- Louise Iara Gomes de Oliveira
- Post-Graduate Program in Nutritional Sciences, Health Sciences Center, Federal University of Paraíba (Universidade Federal da Paraíba - UFPB), Brazil.
| | - Jéssika Rodrigues Clementino
- Post-Graduate Program in Nutritional Sciences, Health Sciences Center, Federal University of Paraíba (Universidade Federal da Paraíba - UFPB), Brazil.
| | - Mateus Kawata Salgaço
- Department of Food and Nutrition, Laboratory of Food Microbiology, School of Pharmaceutical Sciences, São Paulo State University, Brazil
| | - Sônia Paula Alexandrino de Oliveira
- Post-Graduate Program in Nutritional Sciences, Health Sciences Center, Federal University of Paraíba (Universidade Federal da Paraíba - UFPB), Brazil.
| | - Marcos Dos Santos Lima
- Department of Food Technology, Federal Institute of Sertão de Pernambuco, Campus Petrolina, Brazil
| | - Victoria Mesa
- Food and Human Nutrition Research Group, School of Nutrition and Dietetics, Universidad de Antioquia (UdeA), Medellín 050010, Colombia
| | - Evandro Leite de Souza
- Post-Graduate Program in Nutritional Sciences, Health Sciences Center, Federal University of Paraíba (Universidade Federal da Paraíba - UFPB), Brazil.
| | - Celso Gabriel Vinderola
- Department of Biotechnology and Food Technology, Faculty of Chemical Engineering, Universidad Nacional del Litoral
| | - Marciane Magnani
- Post-Graduate Program in Nutritional Sciences, Health Sciences Center, Federal University of Paraíba (Universidade Federal da Paraíba - UFPB), Brazil.
| | - Katia Sivieri
- Department of Food and Nutrition, Laboratory of Food Microbiology, School of Pharmaceutical Sciences, São Paulo State University, Brazil
| |
Collapse
|
10
|
Cesar T, Salgaço MK, Mesa V, Sartoratto A, Sivieri K. Exploring the Association between Citrus Nutraceutical Eriocitrin and Metformin for Improving Pre-Diabetes in a Dynamic Microbiome Model. Pharmaceuticals (Basel) 2023; 16:650. [PMID: 37242433 PMCID: PMC10221435 DOI: 10.3390/ph16050650] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Pre-diabetes is recognized as an altered metabolic state, which precedes type 2 diabetes, and it is associated with great dysfunction of the intestinal microbiota, known as dysbiosis. Natural compounds, capable of reducing blood glucose without side effects and with a beneficial effect on the microbiota, have been studied as substitutes or adjuvants to conventional hypoglycemic agents, such as metformin. In this work, the effect of the nutraceutical Eriomin®, a mixture of citrus flavonoids (eriocitrin, hesperidin, naringin, and didymin), which reduces glycemia and increases glucagon-like peptide-1 (GLP-1) in pre-diabetic patients, was tested in the Simulator of Human Intestinal Microbial Ecosystem (SHIME®), inoculated with pre-diabetic microbiota. After treatment with Eriomin® plus metformin, a significant increase in acetate and butyrate production was observed. Furthermore, sequencing of the 16S rRNA gene of the microorganisms showed that Eriomin® plus metformin stimulated the growth of Bacteroides and Subdoligranulum genera. Bacteroides are the largest fraction of the intestinal microbiota and are potential colonizers of the colon, with some species producing acetic and propionic fatty acids. In addition, Subdoligranulum species are associated with better host glycemic metabolism. In conclusion, Eriomin® associated with metformin improved the composition and metabolism of the intestinal microbiota, suggesting a potential use in pre-diabetes therapy.
Collapse
Affiliation(s)
- Thais Cesar
- Graduate Program in Food, Nutrition and Food Engineering, Campus Araraquara, São Paulo State University (UNESP), Araraquara 14800-060, SP, Brazil; (T.C.)
| | - Mateus Kawata Salgaço
- Graduate Program in Food, Nutrition and Food Engineering, Campus Araraquara, São Paulo State University (UNESP), Araraquara 14800-060, SP, Brazil; (T.C.)
| | - Victoria Mesa
- INSERM, UMR-S 1139 (3PHM), Faculty of Pharmacy, Université Paris Cité, F-75006 Paris, France
- Food and Human Nutrition Research Group, School of Nutrition and Dietetics, Universidad de Antioquia (UdeA), Medellín 050010, Antioquia, Colombia
| | | | - Katia Sivieri
- Graduate Program in Food, Nutrition and Food Engineering, Campus Araraquara, São Paulo State University (UNESP), Araraquara 14800-060, SP, Brazil; (T.C.)
| |
Collapse
|
11
|
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
|
12
|
De Oliveira FL, Salgaço MK, de Oliveira MT, Mesa V, Sartoratto A, Peregrino AM, Ramos WS, Sivieri K. Exploring the Potential of Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 as Promising Psychobiotics Using SHIME. Nutrients 2023; 15:nu15061521. [PMID: 36986251 PMCID: PMC10056475 DOI: 10.3390/nu15061521] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/12/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Psychobiotics are probiotics that have the characteristics of modulating central nervous system (CNS) functions or reconciled actions by the gut-brain axis (GBA) through neural, humoral and metabolic pathways to improve gastrointestinal activity as well as anxiolytic and even antidepressant abilities. The aim of this work was to evaluate the effect of Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 on the gut microbiota of mildly anxious adults using SHIME®. The protocol included a one-week control period and two weeks of treatment with L. helveticus R0052 and B. longum R0175. Ammonia (NH4+), short chain fatty acids (SCFAs), gamma-aminobutyric acid (GABA), cytokines and microbiota composition were determined. Probiotic strains decreased significantly throughout the gastric phase. The highest survival rates were exhibited by L. helveticus R0052 (81.58%; 77.22%) after the gastric and intestinal phase when compared to B. longum (68.80%; 64.64%). At the genus level, a taxonomic assignment performed in the ascending colon in the SHIME® model showed that probiotics (7 and 14 days) significantly (p < 0.005) increased the abundance of Lactobacillus and Olsenella and significantly decreased Lachnospira and Escheria-Shigella. The probiotic treatment (7 and 14 days) decreased (p < 0.001) NH4+ production when compared to the control period. For SCFAs, we observed after probiotic treatment (14 days) an increase (p < 0.001) in acetic acid production and total SCFAs when compared to the control period. Probiotic treatment increased (p < 0.001) the secretion of anti-inflammatory (IL-6 and IL-10) and decreased (p < 0.001) pro-inflammatory cytokines (TNF-alpha) when compared to the control period. The gut-brain axis plays an important role in the gut microbiota, producing SCFAs and GABA, stimulating the production of anti-anxiety homeostasis. The signature of the microbiota in anxiety disorders provides a promising direction for the prevention of mental illness and opens a new perspective for using the psychobiotic as a main actor of therapeutic targets.
Collapse
Affiliation(s)
- Fellipe Lopes De Oliveira
- Graduate Program in Food, Nutrition, and Food Engineering, Campus Araraquara, São Paulo State University (UNESP), Araraquara 14800-060, SP, Brazil
| | - Mateus Kawata Salgaço
- Graduate Program in Food, Nutrition, and Food Engineering, Campus Araraquara, São Paulo State University (UNESP), Araraquara 14800-060, SP, Brazil
| | | | - Victoria Mesa
- Université Paris Cité, INSERM, UMR-S 1139 (3PHM), Faculty of Pharmacy, F-75006 Paris, France
- Food and Human Nutrition Research Group, School of Nutrition and Dietetics, Universidad de Antioquia (UdeA), Medellín 050010, Antioquia, Colombia
| | | | | | - Williams Santos Ramos
- APSEN Farmacêutica, Department of Medical Affairs, Santo Amaro 04753-001, SP, Brazil
| | - Katia Sivieri
- Graduate Program in Food, Nutrition, and Food Engineering, Campus Araraquara, São Paulo State University (UNESP), Araraquara 14800-060, SP, Brazil
- University of Araraquara-UNIARA, Araraquara 14801-320, SP, Brazil
| |
Collapse
|
13
|
Hussain A, Patwekar U, Mongad DS, Shouche YS. Strategizing the human microbiome for small molecules: Approaches and perspectives. Drug Discov Today 2023; 28:103459. [PMID: 36435302 DOI: 10.1016/j.drudis.2022.103459] [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: 02/28/2022] [Revised: 11/03/2022] [Accepted: 11/20/2022] [Indexed: 11/24/2022]
Abstract
Studies of the human microbiome are providing a deeper understanding of its significance to human health, and increasing evidence links the microbiota with several diseases. Nevertheless, the exact mechanisms involved in human-microbe interactions are mostly undefined. The genomic potential of the human microbiome to biosynthesize distinct molecules outmatches its known chemical space, and small-molecule discovery in this context remains in its infancy. The profiling of microbiome-derived small molecules and their contextualization through cause-effect mechanistic studies may provide a better understanding of host-microbe interactions, guide new therapeutic interventions, and modulate microbiome-based therapies. This review describes the advances, approaches, and allied challenges in mining new microbial scaffolds from the human microbiome using genomic, microbe cultivation, and chemical analytic platforms. In the future, the complete biological characterization of a single microbe-derived molecule that has a specific therapeutic application could resolve the current limitations of microbiota-modulating therapies.
Collapse
Affiliation(s)
- Aehtesham Hussain
- NCMR-National Centre for Cell Science (NCCS), Pune, Maharashtra 411007, India.
| | - Umera Patwekar
- NCMR-National Centre for Cell Science (NCCS), Pune, Maharashtra 411007, India
| | - Dattatray S Mongad
- NCMR-National Centre for Cell Science (NCCS), Pune, Maharashtra 411007, India
| | - Yogesh S Shouche
- NCMR-National Centre for Cell Science (NCCS), Pune, Maharashtra 411007, India
| |
Collapse
|
14
|
Functional Fermented Milk with Fruit Pulp Modulates the In Vitro Intestinal Microbiota. Foods 2022; 11:foods11244113. [PMID: 36553855 PMCID: PMC9778618 DOI: 10.3390/foods11244113] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
The effect of putative probiotic fermented milk (FM) with buriti pulp (FMB) or passion fruit pulp (FMPF) or without fruit pulp (FMC) on the microbiota of healthy humans was evaluated. FM formulations were administered into a simulator of the human intestinal microbial ecosystem (SHIME®) to evaluate the viability of lactic acid bacteria (LAB), microbiota composition, presence of short-chain fatty acids (SCFA), and ammonium ions. The probiotic LAB viability in FM was affected by the addition of the fruit pulp. Phocaeicola was dominant in the FMPF and FMB samples; Bifidobacterium was related to FM formulations, while Alistipes was associated with FMPF and FMB, and Lactobacillus and Lacticaseibacillus were predominant in FMC. Trabulsiella was the central element in the FMC, while Mediterraneibacter was the central one in the FMPF and FMB networks. The FM formulations increased the acetic acid, and a remarkably high amount of propionic and butyric acids were detected in the FMB treatment. All FM formulations decreased the ammonium ions compared to the control; FMPF samples stood out for having lower amounts of ammonia. The probiotic FM with fruit pulp boosted the beneficial effects on the intestinal microbiota of healthy humans in addition to increasing SCFA in SHIME® and decreasing ammonium ions, which could be related to the presence of bioactive compounds.
Collapse
|
15
|
Tonon KM, Salgaço MK, Mesa V, Bento Mosquera EM, Tomé TM, Freitas PV, Machado Alencar NM, Sartoratto A, Lazarini T, Sivieri K. Infant formula with 2’-FL + LNnT positively modulates the infant gut microbiome: an in vitro study using Human Intestinal Microbial Ecosystem model. Int Dairy J 2022. [DOI: 10.1016/j.idairyj.2022.105558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
16
|
Giannini C, Mastromauro C, Scapaticci S, Gentile C, Chiarelli F. Role of bile acids in overweight and obese children and adolescents. Front Endocrinol (Lausanne) 2022; 13:1011994. [PMID: 36531484 PMCID: PMC9747777 DOI: 10.3389/fendo.2022.1011994] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/07/2022] [Indexed: 12/05/2022] Open
Abstract
Bile acids (BAs) are amphipathic molecules synthetized in the liver. They are primarily involved in the digestion of nutrients. Apart from their role in dietary lipid absorption, BAs have progressively emerged as key regulators of systemic metabolism and inflammation. In the last decade, it became evident that BAs are particularly important for the regulation of glucose, lipid, and energy metabolism. Indeed, the interest in role of BA in metabolism homeostasis is further increased due to the global public health increase in obesity and related complications and a large number of research postulating that there is a close mutual relationship between BA and metabolic disorders. This strong relationship seems to derive from the role of BAs as signaling molecules involved in the regulation of a wide spectrum of metabolic pathways. These actions are mediated by different receptors, particularly nuclear farnesoid X receptor (FXR) and Takeda G protein coupled receptor 5 (TGR5), which are probably the major effectors of BA actions. These receptors activate transcriptional networks and signaling cascades controlling the expression and activity of genes involved in BA, lipid and carbohydrate metabolism, energy expenditure, and inflammation. The large correlation between BAs and metabolic disorders offers the possibility that modulation of BAs could be used as a therapeutic approach for the treatment of metabolic diseases, including obesity itself. The aim of this review is to describe the main physiological and metabolic actions of BA, focusing on its signaling pathways, which are important in the regulation of metabolism and might provide new BA -based treatments for metabolic diseases.
Collapse
Affiliation(s)
- Cosimo Giannini
- Department of Pediatrics, University of Chieti, Chieti, Italy
| | | | | | | | | |
Collapse
|
17
|
Liu P, Hu T, Kang C, Liu J, Zhang J, Ran H, Zeng X, Qiu S. Research Advances in the Treatment of Allergic Rhinitis by Probiotics. J Asthma Allergy 2022; 15:1413-1428. [PMID: 36238950 PMCID: PMC9552798 DOI: 10.2147/jaa.s382978] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/11/2022] [Indexed: 11/23/2022] Open
Abstract
Allergic rhinitis (AR) impairs the quality of life of patients and reduces the efficiency of social work, it is an increasingly serious public medical and economic problem in the world. Conventional anti-allergic drugs for the treatment of allergic rhinitis (AR) can cause certain side effects, which limit the quality of life of patients. Therefore, it makes sense to look for other forms of treatment. Several studies in recent years have shown that probiotics have shown anti-allergic effects in various mouse and human studies. For example, the application of certain probiotic strains can effectively relieve the typical nasal and ocular symptoms of allergic rhinitis in children and adults, thereby improving the quality of life and work efficiency. At the same time, previous studies in humans and mice have found that probiotics can produce multiple effects, such as reduction of Th2 cell inflammatory factors and/or increase of Th1 cell inflammatory factors, changes in allergy-related immunoglobulins and cell migration, regulate Th1/Th2 balance or restore intestinal microbiota disturbance. For patients with limited activity or allergic rhinitis with more attacks and longer attack duration, oral probiotics have positive effects. The efficacy of probiotics in the prevention and treatment of allergic rhinitis is remarkable, but its specific mechanism needs further study. This review summarizes the research progress of probiotics in the treatment of allergic rhinitis in recent years.
Collapse
Affiliation(s)
- Peng Liu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zunyi, People’s Republic of China
| | - Tianyong Hu
- Department of Otolaryngology, Longgang E.N.T Hospital & Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, People’s Republic of China
| | - Chenglin Kang
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zunyi, People’s Republic of China
| | - Jiangqi Liu
- Department of Otolaryngology, Longgang E.N.T Hospital & Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, People’s Republic of China
| | - Jin Zhang
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zunyi, People’s Republic of China
| | - Hong Ran
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zunyi, People’s Republic of China
| | - Xianhai Zeng
- Department of Otolaryngology, Longgang E.N.T Hospital & Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, People’s Republic of China
| | - Shuqi Qiu
- Department of Otolaryngology, Longgang E.N.T Hospital & Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, People’s Republic of China
| |
Collapse
|
18
|
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: 3.3] [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
|
19
|
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: 31] [Impact Index Per Article: 10.3] [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
|
20
|
Barros de Medeiros VP, Salgaço MK, Pimentel TC, Rodrigues da Silva TC, Sartoratto A, Lima MDS, Sassi CFDC, Mesa V, Magnani M, Sivieri K. Spirulina platensis biomass enhances the proliferation rate of Lactobacillus acidophilus 5 (La-5) and combined with La-5 impact the gut microbiota of medium-age healthy individuals through an in vitro gut microbiome model. Food Res Int 2022; 154:110880. [PMID: 35337549 DOI: 10.1016/j.foodres.2021.110880] [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: 10/06/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 11/16/2022]
Abstract
This study first evaluated the stimulatory effect of S. platensis biomass on the growth of L. acidophilus and the metabolic activity during fermentation (37 °C, 72 h) in a culture medium. The results demonstrated a higher impact of S. platensis biomass than fructooligosaccharide (FOS), an established prebiotic. Higher L. acidophilus proliferation rates and metabolic activity were observed (lower pH values and higher concentrations of acetic, lactic, and propionic acids) in the presence of S. platensis. Then, we evaluated the effects of the S. platensis biomass (1.5 g, twice a day, 5 days) in association with L. acidophilus (106 CFU/g) on the gut microbiota composition of medium-age healthy individuals through the Simulator of Human Intestinal Microbial Ecosystem (SHIME®) and measurement of metabolites. L. acidophilus (La5) and L. acidophilus + S. platensis (Spi-La5) could positively modulate the intestinal microbiota. The administration of La5 resulted in increases in Bacteroides, Megasphaera, Lactobacillus, and Parabacteroides genus abundance, with a consequent decrease in ammonium ions. The administration of Spi-La5 increased the abundance of the genus Erysipelatoclostridium, Roseburia, Enterococcus, Bifidobacterium, Coriobacteriaceae UCG-003, Enterobacter, and Paraclostridium. The results demonstrate that the intestinal microbiota was differently modified by administrating La5 and Spi-La5 and indicate the latter as an alternative for microbiota positive modulation in healthy individuals.
Collapse
Affiliation(s)
| | - Mateus Kawata Salgaço
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | | | | | - Adilson Sartoratto
- Pluridisciplinary Center for Chemical, Biological and Agricultural Research (CPQBA), UNICAMP, Brazil
| | - Marcos Dos Santos Lima
- Departament of Food Technology, Federal Institute of Sertão Pernambucano, Pernambuco, Brazil
| | | | - Victoria Mesa
- Food and Human Nutrition Research Group, University of Antioquia, Medellín, Colombia
| | - Marciane Magnani
- Department of Food Engineering, Center of Technology, Federal University of Paraíba, PB, Brazil.
| | - Katia Sivieri
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| |
Collapse
|
21
|
Wang T, Chen W, Shao Y, Liu J, Tu Z. Ultrasound Improved the Non-Covalent Interaction of β-Lactoglobulin with Luteolin: Regulating Human Intestinal Microbiota and Conformational Epitopes Reduced Allergy Risks. Foods 2022; 11:foods11070988. [PMID: 35407075 PMCID: PMC8997858 DOI: 10.3390/foods11070988] [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: 02/28/2022] [Revised: 03/21/2022] [Accepted: 03/24/2022] [Indexed: 11/27/2022] Open
Abstract
The present study aims to investigate the effects of ultrasound on the non-covalent interaction of β-lactoglobulin (β-LG) and luteolin (LUT) and to investigate the relationship between allergenicity and human intestinal microbiota. After treatment, the conformational structures of β-LG were changed, which reflected by the decrease in α-helix content, intrinsic fluorescence intensity and surface hydrophobicity, whereas the β-sheet content increased. Molecular docking studies revealed the non-covalent interaction of β-LG and LUT by hydrogen bond, van der Walls bond and hydrophobic bond. β-LG-LUT complex treated by ultrasound has a lower IgG/IgE binding ability and inhibits the allergic reaction of KU812 cells, depending on the changes in the conformational epitopes of β-LG. Meanwhile, the β-LG-LUT complex affected the composition of human intestinal microbiota, such as the relative abundance of Bifidobacterium and Prevotella. Therefore, ultrasound improved the non-covalent interaction of β-LG with LUT, and the reduction in allergenicity of β-LG depends on conformational epitopes and human intestinal microbiota changes.
Collapse
Affiliation(s)
- Titi Wang
- College of Life Science, National R & D Center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang 330022, China; (T.W.); (W.C.); (Y.S.); (J.L.)
| | - Wenmei Chen
- College of Life Science, National R & D Center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang 330022, China; (T.W.); (W.C.); (Y.S.); (J.L.)
| | - Yanhong Shao
- College of Life Science, National R & D Center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang 330022, China; (T.W.); (W.C.); (Y.S.); (J.L.)
| | - Jun Liu
- College of Life Science, National R & D Center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang 330022, China; (T.W.); (W.C.); (Y.S.); (J.L.)
| | - Zongcai Tu
- College of Life Science, National R & D Center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang 330022, China; (T.W.); (W.C.); (Y.S.); (J.L.)
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
- Correspondence: ; Tel.: +86-791-8812-1868; Fax: +86-791-8830-5938
| |
Collapse
|
22
|
Zhang S, Dang Y. Roles of gut microbiota and metabolites in overweight and obesity of children. Front Endocrinol (Lausanne) 2022; 13:994930. [PMID: 36157438 PMCID: PMC9492854 DOI: 10.3389/fendo.2022.994930] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
The prevalence of overweight and obesity in children and adolescents is an increasing public health problem. Pediatric overweight and obesity result from multiple factors, including genetic background, diet, and lifestyle. In addition, the gut microbiota and their metabolites play crucial roles in the progression of overweight and obesity of children. Therefore, we reviewed the roles of gut microbiota in overweight/obese children. The relationship between pediatric overweight/obesity and gut metabolites, such as short-chain fatty acids, medium-chain fatty acids, amino acids, amines, and bile acids, are also summarized. Targeting gut microbiota and metabolites might be a promising strategy for interventions aimed at reducing pediatric overweight/obesity.
Collapse
Affiliation(s)
- Shengan Zhang
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanqi Dang
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Yanqi Dang, ,
| |
Collapse
|
23
|
Zhou J, Sun S, Luan S, Xiao X, Yang Y, Mao C, Chen L, Zeng X, Zhang Y, Yuan Y. Gut Microbiota for Esophageal Cancer: Role in Carcinogenesis and Clinical Implications. Front Oncol 2021; 11:717242. [PMID: 34733778 PMCID: PMC8558403 DOI: 10.3389/fonc.2021.717242] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 09/24/2021] [Indexed: 02/05/2023] Open
Abstract
Esophageal cancer (EC) is a common malignant tumor of the upper digestive tract. The microbiota in the digestive tract epithelium comprises a large number of microorganisms that adapt to the immune defense and interact with the host to form symbiotic networks, which affect many physiological processes such as metabolism, tissue development, and immune response. Reports indicate that there are microbial compositional changes in patients with EC, which provides an important opportunity to advance clinical applications based on findings on the gut microbiota. For example, microbiota detection can be used as a biomarker for screening and prognosis, and microorganism levels can be adjusted to treat cancer and decrease the adverse effects of treatment. This review aims to provide an outline of the gut microbiota in esophageal neoplasia, including the mechanisms involved in microbiota-related carcinogenesis and the prospect of utilizing the microbiota as EC biomarkers and treatment targets. These findings have important implications for translating the use of gut microbiota in clinical applications.
Collapse
Affiliation(s)
- Jianfeng Zhou
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Shangwei Sun
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Siyuan Luan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Xiao
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yushang Yang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Chengyi Mao
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Longqi Chen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxi Zeng
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yonggang Zhang
- Department of Periodical Press, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Yuan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Role of Food Antioxidants in Modulating Gut Microbial Communities: Novel Understandings in Intestinal Oxidative Stress Damage and Their Impact on Host Health. Antioxidants (Basel) 2021; 10:antiox10101563. [PMID: 34679698 PMCID: PMC8533511 DOI: 10.3390/antiox10101563] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/21/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023] Open
Abstract
Dietary components have an important role on the structure and function of host gut microbial communities. Even though, various dietary components, such as carbohydrates, fats, proteins, fibers, and vitamins, have been studied in depth for their effect on gut microbiomes, little attention has been paid regarding the impact of several food antioxidants on the gut microbiome. The long-term exposure to reactive oxygen species (ROS) can cause microbial dysbiosis which leads to numerous intestinal diseases such as microbiota dysbiosis, intestinal injury, colorectal cancers, enteric infections, and inflammatory bowel diseases. Recently, it has been shown that the food derived antioxidant compounds might protect the host from intestinal oxidative stress via modulating the composition of beneficial microbial species in the gut. The present review summarizes the impact of food antioxidants including antioxidant vitamins, dietary polyphenols, carotenoids, and bioactive peptides on the structure as well as function of host gut microbial communities. Several in vitro, animal model, and clinical studies indicates that food antioxidants might modify the host gut microbial communities and their health status. However, still further clarification is needed as to whether changes in certain microbial species caused by food additives may lead to changes in metabolism and immune function.
Collapse
|