1
|
Srivastava A, Rastogi A, Jaswal AS, Sahu JK, Agarwal GP, Mishra S. Nanofiltration-based purification process for whole-cell transformed prebiotic galactooligosaccharides. Bioprocess Biosyst Eng 2025:10.1007/s00449-025-03132-6. [PMID: 39903231 DOI: 10.1007/s00449-025-03132-6] [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: 11/22/2024] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
The enrichment of galactooligosaccharides (GOS), synthesized by whole cells of Kluyveromyces marxianus 3551 in a 5.0-L bioreactor, was investigated in this study. The synthesized sugar mixture containing 17.89% (w/w of total carbohydrates) of GOS with 15.57% (w/w of total carbohydrates) of lactose, and 66.54% (w/w of total carbohydrates) monosaccharides as impurities, was subjected to nanofiltration for enrichment of GOS. Three distinct spiral wound membranes, namely, NFPS-01(polysulfone), NFCA-02 (cellulose acetate), and NFPES-03 (polyethersulfone) were tested out of which the NFPES-03 performed the best for fractionation of the GOS mixture. The polyethersulphone membrane (cut-off 400-1000 Da) was evaluated at 30 ℃ and 50 ℃, at different transmembrane pressures or TMP (15, 20, 25 bar) and a combination of high temperature (50 ℃) and low pressure (15 bar) gave the greatest difference in the trisaccharide and disaccharide/monosaccharide rejection percentages, resulting in enrichment of GOS. An analysis of the sugar concentrations in the retentate samples by high-performance liquid chromatography indicated the percentage recovery of GOS in the integrated process to be 88.8%. Measurement of the growth profile of several microbes on the nano-filtered GOS demonstrated its effectiveness as a prebiotic source.
Collapse
Affiliation(s)
- Anita Srivastava
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, Hauz-Khas, New-Delhi, 110016, India.
- Food Bioprocess Engineering Research Laboratory, Centre for Rural Development and Technology, Indian Institute of Technology Delhi, Hauz-Khas, New-Delhi, 110016, India.
| | - Arjun Rastogi
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, Hauz-Khas, New-Delhi, 110016, India
| | - Avijeet S Jaswal
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, Hauz-Khas, New-Delhi, 110016, India
| | - Jatindra K Sahu
- Food Bioprocess Engineering Research Laboratory, Centre for Rural Development and Technology, Indian Institute of Technology Delhi, Hauz-Khas, New-Delhi, 110016, India
| | - Gopal P Agarwal
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, Hauz-Khas, New-Delhi, 110016, India
| | - Saroj Mishra
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, Hauz-Khas, New-Delhi, 110016, India
| |
Collapse
|
2
|
Al-Wraikat M, Zhang L, Li L, Abubaker MA, Liu Y. Recent advances in wolfberry polysaccharides and whey protein-based biopolymers for regulating the diversity of gut microbiota and its mechanism: A review. Int J Biol Macromol 2024; 281:136401. [PMID: 39383924 DOI: 10.1016/j.ijbiomac.2024.136401] [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: 02/08/2024] [Revised: 09/11/2024] [Accepted: 10/05/2024] [Indexed: 10/11/2024]
Abstract
Imbalances in gut microbiota diversity are associated with various health issues, including obesity and related disorders. There is a growing interest in developing synergistic biopolymers based on wolfberry polysaccharides and whey protein to address these problems due to their potential health benefits. This review explores recent advances in understanding how functional foods based on Lycium barbarum polysaccharides (LBP) and whey protein (WP) influence gut microbiota diversity and their underlying mechanisms. We examine the impact of these biopolymers on microbial composition and functionality, focusing on their roles in improving health by regulating gut microbiota. The combined effects of WP and LBP significantly enhance gut microbiome metabolic activities and taxonomic diversity, offering promising avenues for treating obesity and related disorders.
Collapse
Affiliation(s)
- Majida Al-Wraikat
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, Shaanxi, China
| | - Lan Zhang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, Shaanxi, China
| | - Linqiang Li
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, Shaanxi, China
| | - Mohamed Aamer Abubaker
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, Shaanxi, China
| | - Yongfeng Liu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, Shaanxi, China.
| |
Collapse
|
3
|
Yu X, Peng X, Liu F, Li Y, Yan J, Li L. Distinguishing α/β-linkages and linkage positions of disaccharides in galactooligosaccharides through mass fragmentation and liquid retention behaviour. Food Chem 2024; 456:139968. [PMID: 38861865 DOI: 10.1016/j.foodchem.2024.139968] [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: 02/20/2024] [Revised: 05/31/2024] [Accepted: 06/02/2024] [Indexed: 06/13/2024]
Abstract
Galactooligosaccharides (GOS) are important prebiotics with function closely related to their structure. However, a comprehensive overview of the structure-function relationship is still limited due to the challenge in characterizing multiple isomers in GOS. This study presents a strategy of combining both hydrophilic interaction liquid chromatography (HILIC) retention time and tandem mass spectrometry (MS/MS) fragmentation pattern to distinguish α/β-linkages and linkage positions of disaccharide isomers in GOS through HILIC-MS/MS analysis. The results indicated that the ratio of m/z 203.0524 to m/z 365.1054 could distinguish α/β-linkages, while the ratios of m/z 347.0947 to m/z 365.1054, m/z 245.0642 to m/z 365.1054 and HILIC retention time could distinguish (1 → 2), (1 → 3), (1 → 4) and (1 → 6) linkages. The above rules enabled effective characterization of disaccharides in GOS-containing food samples, including milk powder, rice flour, drink, yogurt. This method can be used in the quality control of GOS and future research on the structure-specific health effects of GOS.
Collapse
Affiliation(s)
- Xiangying Yu
- Engineering Research Center of Health Food Design & Nutrition Regulation, Dongguan Key Laboratory of Typical Food Precision Design, China National Light Industry Key Laboratory of Healthy Food Development and Nutrition Regulation, School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China.
| | - Xueying Peng
- Engineering Research Center of Health Food Design & Nutrition Regulation, Dongguan Key Laboratory of Typical Food Precision Design, China National Light Industry Key Laboratory of Healthy Food Development and Nutrition Regulation, School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China
| | - Fengyuan Liu
- Engineering Research Center of Health Food Design & Nutrition Regulation, Dongguan Key Laboratory of Typical Food Precision Design, China National Light Industry Key Laboratory of Healthy Food Development and Nutrition Regulation, School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China.
| | - Yuting Li
- Engineering Research Center of Health Food Design & Nutrition Regulation, Dongguan Key Laboratory of Typical Food Precision Design, China National Light Industry Key Laboratory of Healthy Food Development and Nutrition Regulation, School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China
| | - Jingkun Yan
- Engineering Research Center of Health Food Design & Nutrition Regulation, Dongguan Key Laboratory of Typical Food Precision Design, China National Light Industry Key Laboratory of Healthy Food Development and Nutrition Regulation, School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China.
| | - Lin Li
- Engineering Research Center of Health Food Design & Nutrition Regulation, Dongguan Key Laboratory of Typical Food Precision Design, China National Light Industry Key Laboratory of Healthy Food Development and Nutrition Regulation, School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China.
| |
Collapse
|
4
|
Hu Y, Aljumaah MR, Azcarate-Peril MA. Galacto-Oligosaccharides and the Elderly Gut: Implications for Immune Restoration and Health. Adv Nutr 2024; 15:100263. [PMID: 38897384 PMCID: PMC11292246 DOI: 10.1016/j.advnut.2024.100263] [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: 02/12/2024] [Revised: 04/23/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024] Open
Abstract
The increasing prevalence of noncommunicable diseases in the aging population has been correlated with a decline in innate and adaptive immune responses; hence, it is imperative to identify approaches to improve immune function, prevent related disorders, and reduce or treat age-associated health complications. Prebiotic supplementation is a promising approach to modulate the gut microbiome and immune system, offering a potential strategy to maintain the integrity of immune function in older individuals. This review summarizes the current research on prebiotic galacto-oligosaccharide (GOS) immunomodulatory mechanisms mediated by bacterial-derived metabolites, including short-chain fatty acids and secondary bile acids, to maintain immune homeostasis. The potential applications of GOS as immunotherapy for age-related disease prevention in older individuals are also highlighted. This aligns with the global shift toward proactive healthcare and emphasizes the significance of early intervention in directing an individual's health trajectory.
Collapse
Affiliation(s)
- Yunan Hu
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, United States; UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Mashael R Aljumaah
- UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina, Chapel Hill, NC, United States; Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, United States; Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Maria Andrea Azcarate-Peril
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, United States; UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina, Chapel Hill, NC, United States.
| |
Collapse
|
5
|
Ong MS, Sordillo JE, Dahlin A, McGeachie M, Tantisira K, Wang AL, Lasky-Su J, Brilliant M, Kitchner T, Roden DM, Weiss ST, Wu AC. Machine Learning Prediction of Treatment Response to Inhaled Corticosteroids in Asthma. J Pers Med 2024; 14:246. [PMID: 38540988 PMCID: PMC10970828 DOI: 10.3390/jpm14030246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Although inhaled corticosteroids (ICS) are the first-line therapy for patients with persistent asthma, many patients continue to have exacerbations. We developed machine learning models to predict the ICS response in patients with asthma. METHODS The subjects included asthma patients of European ancestry (n = 1371; 448 children; 916 adults). A genome-wide association study was performed to identify the SNPs associated with ICS response. Using the SNPs identified, two machine learning models were developed to predict ICS response: (1) least absolute shrinkage and selection operator (LASSO) regression and (2) random forest. RESULTS The LASSO regression model achieved an AUC of 0.71 (95% CI 0.67-0.76; sensitivity: 0.57; specificity: 0.75) in an independent test cohort, and the random forest model achieved an AUC of 0.74 (95% CI 0.70-0.78; sensitivity: 0.70; specificity: 0.68). The genes contributing to the prediction of ICS response included those associated with ICS responses in asthma (TPSAB1, FBXL16), asthma symptoms and severity (ABCA7, CNN2, PTRN3, and BSG/CD147), airway remodeling (ELANE, FSTL3), mucin production (GAL3ST), leukotriene synthesis (GPX4), allergic asthma (ZFPM1, SBNO2), and others. CONCLUSIONS An accurate risk prediction of ICS response can be obtained using machine learning methods, with the potential to inform personalized treatment decisions. Further studies are needed to examine if the integration of richer phenotype data could improve risk prediction.
Collapse
Affiliation(s)
- Mei-Sing Ong
- PRecisiOn Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care, Boston, MA 02215, USA; (J.E.S.); (A.C.W.)
| | - Joanne E. Sordillo
- PRecisiOn Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care, Boston, MA 02215, USA; (J.E.S.); (A.C.W.)
| | - Amber Dahlin
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (A.D.); (M.M.); (A.L.W.); (J.L.-S.); (S.T.W.)
| | - Michael McGeachie
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (A.D.); (M.M.); (A.L.W.); (J.L.-S.); (S.T.W.)
| | - Kelan Tantisira
- Division of Pediatric Respiratory Medicine, Department of Pediatrics, University of California San Diego and Rady Children’s Hospital, San Diego, CA 92123, USA;
| | - Alberta L. Wang
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (A.D.); (M.M.); (A.L.W.); (J.L.-S.); (S.T.W.)
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (A.D.); (M.M.); (A.L.W.); (J.L.-S.); (S.T.W.)
| | - Murray Brilliant
- Marshfield Clinic Research Institute, Marshfield, WI 54449, USA; (M.B.); (T.K.)
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Terrie Kitchner
- Marshfield Clinic Research Institute, Marshfield, WI 54449, USA; (M.B.); (T.K.)
| | - Dan M. Roden
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Scott T. Weiss
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (A.D.); (M.M.); (A.L.W.); (J.L.-S.); (S.T.W.)
| | - Ann Chen Wu
- PRecisiOn Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care, Boston, MA 02215, USA; (J.E.S.); (A.C.W.)
| |
Collapse
|
6
|
Yin Z, Liu X, Guo L, Ren M, Kang W, Ma C, Waterhouse GIN, Sun-Waterhouse D. The potential of dietary fiber in building immunity against gastrointestinal and respiratory disorders. Crit Rev Food Sci Nutr 2023; 64:13318-13336. [PMID: 37837407 DOI: 10.1080/10408398.2023.2266462] [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: 10/16/2023]
Abstract
The numerous health benefits of dietary fibers (DFs) justify their inclusion in human diets and biomedical products. Given the short- and long-term human impacts of the COVID-19 virus on human health, the potential of DFs in building immunity against gastrointestinal and respiratory disorders is currently receiving high attention. This paper reviews the physicochemical properties of DFs, together with their immune functions and effects on the gastrointestinal tract and respiratory system mainly based on research in the last ten years. Possible modes of action of DFs in promoting health, especially building immunity, are explored. We seek to highlight the importance of understanding the exact physical and chemical characteristics and molecular behaviors of DFs in providing specific immune function. This review provides a perspective beyond the existing recognition of DFs' positive effects on human health, and offers a theoretical framework for the development of special DFs components and their application in functional foods and other therapeutic products against gastrointestinal and respiratory disorders. DFs enhance immunity from gastrointestinal and respiratory diseases to promote host health.
Collapse
Affiliation(s)
- Zhenhua Yin
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Comprehensive Utilization of Edible and Medicinal Plant Resources Engineering Technology Research Center, Huanghe Science and Technology College, Zhengzhou, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Xiaopeng Liu
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Lin Guo
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Mengjie Ren
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Wenyi Kang
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Changyang Ma
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | | | | |
Collapse
|
7
|
Gu M, Yin W, Zhang J, Yin J, Tang X, Ling J, Tang Z, Yin W, Wang X, Ni Q, Zhu Y, Chen T. Role of gut microbiota and bacterial metabolites in mucins of colorectal cancer. Front Cell Infect Microbiol 2023; 13:1119992. [PMID: 37265504 PMCID: PMC10229905 DOI: 10.3389/fcimb.2023.1119992] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/03/2023] [Indexed: 06/03/2023] Open
Abstract
Colorectal cancer (CRC) is a major health burden, accounting for approximately 10% of all new cancer cases worldwide. Accumulating evidence suggests that the crosstalk between the host mucins and gut microbiota is associated with the occurrence and development of CRC. Mucins secreted by goblet cells not only protect the intestinal epithelium from microorganisms and invading pathogens but also provide a habitat for commensal bacteria. Conversely, gut dysbiosis results in the dysfunction of mucins, allowing other commensals and their metabolites to pass through the intestinal epithelium, potentially triggering host responses and the subsequent progression of CRC. In this review, we summarize how gut microbiota and bacterial metabolites regulate the function and expression of mucin in CRC and novel treatment strategies for CRC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xiangjun Wang
- *Correspondence: Xiangjun Wang, ; Qing Ni, ; Yunxiang Zhu, ; Tuo Chen,
| | - Qing Ni
- *Correspondence: Xiangjun Wang, ; Qing Ni, ; Yunxiang Zhu, ; Tuo Chen,
| | - Yunxiang Zhu
- *Correspondence: Xiangjun Wang, ; Qing Ni, ; Yunxiang Zhu, ; Tuo Chen,
| | - Tuo Chen
- *Correspondence: Xiangjun Wang, ; Qing Ni, ; Yunxiang Zhu, ; Tuo Chen,
| |
Collapse
|
8
|
Chen X, de Vos P. Structure-function relationship and impact on the gut-immune barrier function of non-digestible carbohydrates and human milk oligosaccharides applicable for infant formula. Crit Rev Food Sci Nutr 2023; 64:8325-8345. [PMID: 37035930 DOI: 10.1080/10408398.2023.2199072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Human milk oligosaccharides (hMOs) in mothers' milk play a crucial role in guiding the colonization of microbiota and gut-immune barrier development in infants. Non-digestible carbohydrates (NDCs) such as synthetic single hMOs, galacto-oligosaccharides (GOS), inulin-type fructans and pectin oligomers have been added to infant formula to substitute some hMOs' functions. HMOs and NDCs can modulate the gut-immune barrier, which is a multiple-layered functional unit consisting of microbiota, a mucus layer, gut epithelium, and the immune system. There is increasing evidence that the structures of the complex polysaccharides may influence their efficacy in modulating the gut-immune barrier. This review focuses on the role of different structures of individual hMOs and commonly applied NDCs in infant formulas in (i) direct regulation of the gut-immune barrier in a microbiota-independent manner and in (ii) modulation of microbiota composition and microbial metabolites of these polysaccharides in a microbiota-dependent manner. Both have been shown to be essential for guiding the development of an adequate immune barrier, but the effects are very dependent on the structural features of hMO or NDC. This knowledge might lead to tailored infant formulas for specific target groups.
Collapse
Affiliation(s)
- Xiaochen Chen
- Immunoendocrinology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Paul de Vos
- Immunoendocrinology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
9
|
Nie C, Xie X, Liu H, Yuan X, Ma Q, Tu A, Zhang M, Chen Z, Li J. Galactooligosaccharides ameliorate dietary advanced glycation end product-induced intestinal barrier damage in C57BL/6 mice by modulation of the intestinal microbiome. Food Funct 2023; 14:845-856. [PMID: 36537141 DOI: 10.1039/d2fo02959f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Advanced glycation end products (AGEs) are increasingly recognized as potentially pathogenic components of processed foods, and long-term consumption of dietary AGEs triggers disruption of the intestinal barrier integrity and increases the risk of chronic diseases. Galactooligosaccharides (GOS) as prebiotics can modulate the intestinal microbiota and improve the intestinal barrier integrity. In this study, we aimed to investigate whether GOS could ameliorate the intestinal barrier damage induced by AGEs. The results showed an increased number of goblet cells (AGEs vs. H-GOS, 133.4 vs. 174.7, p < 0.05) and neutral mucin area (PAS positive area, 7.29% vs. 10.05%, p < 0.05). Upregulated expressions of occludin and claudin-1 and improved intestinal barrier integrity were observed in the H-GOS group. Using 16S rRNA sequencing analysis, we found that GOS significantly reduced the high enrichment of Akkermansia (16.95% vs. 1.29%, p < 0.05) induced by dietary AGEs while increasing the content of short-chain fatty acids. Fecal microbiota transplantation (FMT) showed that AGE-induced damage to the intestinal mucus barrier was reversed in the H-GOS transplanted group. Collectively, GOS ameliorated dietary AGE-induced intestinal barrier damage by reversing the dysregulated state of the intestinal microbiota. Our study lays the foundation for further research on dietary guidelines for populations with high AGE diets.
Collapse
Affiliation(s)
- Chenxi Nie
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| | - Xiaoqing Xie
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| | - Huicui Liu
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| | - Xiaojin Yuan
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| | - Qingyu Ma
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| | - Aobai Tu
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| | - Min Zhang
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| | - Zhifei Chen
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| | - Juxiu Li
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| |
Collapse
|
10
|
The Milk Active Ingredient, 2'-Fucosyllactose, Inhibits Inflammation and Promotes MUC2 Secretion in LS174T Goblet Cells In Vitro. Foods 2023; 12:foods12010186. [PMID: 36613400 PMCID: PMC9818439 DOI: 10.3390/foods12010186] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 01/03/2023] Open
Abstract
In several mice inflammatory models, human milk oligosaccharides (HMOs) were shown to protect the intestinal barrier by promoting mucin secretion and suppressing inflammation. However, the functions of the individual HMOs in enhancing mucin expression in vivo have not been compared, and the related mechanisms are not yet to be clarified. In this study, we investigated the modulatory effects of 2′-fucosyllactose (2′-FL), 3′-sialyllactose (3′-SL), galacto-oligosaccharide (GOS) and lactose (Lac) on goblet cells’ functions in vitro. The appropriate dosage of the four chemicals was assessed in LS174T cells using the CCK-8 method. Then they were supplemented into a homeostasis and inflammatory environment to further investigate their effects under different conditions. Mucin secretion-related genes, including mucin 2 (MUC2), trefoil factor family 3 (TFF3), resistin-like β (RETNLB), carbohydrate sulfotransferase 5 (CHST5) and galactose-3-O-sulfotransferase 2 (GAL3ST2), in LS174T cells were detected using quantitative RT-qPCR. The results showed that 2′-FL (2.5 mg/mL, 72 h) was unable to increase MUC2 secretion in a steady-state condition. Comparatively, it exhibited a greater ability to improve mucin secretion under an inflammatory condition compared with GOS, demonstrated by a significant increase in TFF3 and CHST5 mRNA expression levels (p > 0.05). However, 3′-SL and Lac exhibited no effects on mucin secretion. To further investigate the underlying mechanism via which 2′-FL enhanced goblet cells’ secretion function, the NOD-like receptor family pyrin domain containing 6 (NLRP6) gene, which is closely related to MUC2 secretion, was silenced using the siRNA method. After silencing the NLRP6 gene, the mRNA expression levels of MUC2, TFF3 and CHST5 in the (2′-FL + tumor necrosis factor α (TNF-α) + NLRP6 siRNA) group were significantly decreased compared with the (2′-FL + TNF-α) group (p > 0.05), indicating that NLRP6 was essential for MUC2 expression in goblet cells. We further found that 2′-FL could significantly decrease toll-like receptor 4 (TLR4, p < 0.05), myeloid differential protein-88 (MyD88, p < 0.05) and nuclear factor kappa-B (NF-κB, p < 0.05) levels in LS174T inflammatory cells, even when the NLRP6 was silenced. Altogether, these results indicated that in goblet cells, 2′-FL exerts its function via multiple processes, i.e., by promoting mucin secretion through NLRP6 and suppressing inflammation by inhibiting the TLR4/MyD88/NF-κB pathway.
Collapse
|
11
|
Cao C, Wang Z, Gong G, Huang W, Huang L, Song S, Zhu B. Effects of Lycium barbarum Polysaccharides on Immunity and Metabolic Syndrome Associated with the Modulation of Gut Microbiota: A Review. Foods 2022; 11:3177. [PMID: 37430929 PMCID: PMC9602392 DOI: 10.3390/foods11203177] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/30/2022] [Accepted: 09/30/2022] [Indexed: 11/17/2022] Open
Abstract
Lycium barbarum polysaccharides (LBPs) have attracted increasing attention due to their multiple pharmacological activities and physiological functions. Recently, both in vitro and in vivo studies have demonstrated that the biological effects of dietary LBPs are related to the regulation of gut microbiota. Supplementation with LBPs could modulate the composition of microbial communities, and simultaneously influence the levels of active metabolites, thus exerting their beneficial effects on host health. Interestingly, LBPs with diverse chemical structures may enrich or reduce certain specific intestinal microbes. The present review summarizes the extraction, purification, and structural types of LBPs and the regulation effects of LBPs on the gut microbiome and their derived metabolites. Furthermore, the health promoting effects of LBPs on host bidirectional immunity (e.g., immune enhancement and immune inflammation suppression) and metabolic syndrome (e.g., obesity, type 2 diabetes, and nonalcoholic fatty liver disease) by targeting gut microbiota are also discussed based on their structural types. The contents presented in this review might help to better understand the health benefits of LBPs targeting gut microbiota and provide a scientific basis to further clarify the structure-function relationship of LBPs.
Collapse
Affiliation(s)
- Cui Cao
- Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
- National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, China
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi’an 710069, China
| | - Zhongfu Wang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi’an 710069, China
| | - Guiping Gong
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi’an 710069, China
| | - Wenqi Huang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi’an 710069, China
| | - Linjuan Huang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi’an 710069, China
| | - Shuang Song
- Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
- National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, China
| | - Beiwei Zhu
- Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
- National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
12
|
Wang G, Wang H, Jin Y, Xiao Z, Umar Yaqoob M, Lin Y, Chen H, Wang M. Galactooligosaccharides as a protective agent for intestinal barrier and its regulatory functions for intestinal microbiota. Food Res Int 2022; 155:111003. [DOI: 10.1016/j.foodres.2022.111003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 12/21/2021] [Accepted: 01/08/2022] [Indexed: 11/04/2022]
|
13
|
Morales-Ferré C, Azagra-Boronat I, Massot-Cladera M, Tims S, Knipping K, Garssen J, Knol J, Franch À, Castell M, Pérez-Cano FJ, Rodríguez-Lagunas MJ. Preventive Effect of a Postbiotic and Prebiotic Mixture in a Rat Model of Early Life Rotavirus Induced-Diarrhea. Nutrients 2022; 14:nu14061163. [PMID: 35334820 PMCID: PMC8954028 DOI: 10.3390/nu14061163] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 02/06/2023] Open
Abstract
Rotavirus (RV) is the main cause of gastroenteritis in children. Prebiotics and, more recently, postbiotics are used for preventing and treating gastrointestinal infections. The aim of this study was to analyze the effects of a LactofidusTM, short-chain galacto-oligosaccharides (scGOS) and long-chain fructo-oligosaccharides (lcFOS) mixture, and their combination on RV infection, in a rat model, for early life diarrhea. Fifteen litters of suckling rats were intragastrically administered daily with the vehicle, the prebiotic mixture, the postbiotic or the combination. The RV was inoculated on day 5 and then fecal samples were clinically evaluated daily. Viral shedding, intestinal permeability assay, in vitro blocking assay, immunoglobulin profiles, and anti-RV response were assessed at day 8 and 16 of life. Cecal microbiota composition, intestinal gene expression, and short chain fatty acids (SCFAs) were analyzed at day 16. The incidence and severity of diarrhea were significantly reduced by all the supplementations. Moreover, they showed blocking activity, changes in the immunoglobulin profiles, in gut microbiota, and in the intestinal gene expression. The prebiotic mixture reduced gut permeability and changed the SCFA profile, whereas the postbiotic enhanced the expression of Toll-like receptors (TLRs). The combination preserved most of the individual observed effects, and furthermore, complementary effects, such as an increase in white blood cells and lymphocytes recruitment, as well as upregulation of TLR7 and TLR9 gene expression.
Collapse
Affiliation(s)
- Carla Morales-Ferré
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Ignasi Azagra-Boronat
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Malén Massot-Cladera
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Sebastian Tims
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
| | - Karen Knipping
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Johan Garssen
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Jan Knol
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Laboratory of Microbiology, Wageningen University, 6708 PB Wageningen, The Netherlands
| | - Àngels Franch
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Francisco J. Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
- Correspondence: ; Tel.: +34-934-024-505
| | - María J. Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|
14
|
Ambrogi V, Bottacini F, Mac Sharry J, van Breen J, O'Keeffe E, Walsh D, Schoemaker B, Cao L, Kuipers B, Lindner C, Jimeno ML, Doyagüez EG, Hernandez-Hernandez O, Moreno FJ, Schoterman M, van Sinderen D. Bifidobacterial β-Galactosidase-Mediated Production of Galacto-Oligosaccharides: Structural and Preliminary Functional Assessments. Front Microbiol 2021; 12:750635. [PMID: 34777303 PMCID: PMC8581567 DOI: 10.3389/fmicb.2021.750635] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
In the current study the ability of four previously characterized bifidobacterial β-galactosidases (designated here as BgaA, BgaC, BgaD, and BgaE) to produce galacto-oligosaccharides (GOS) was optimized. Of these enzymes, BgaA and BgaE were found to be promising candidates for GOS production (and the corresponding GOS mixtures were called GOS-A and GOS-E, respectively) with a GOS concentration of 19.0 and 40.3% (of the initial lactose), respectively. GOS-A and GOS-E were partially purified and structurally characterized. NMR analysis revealed that the predominant (non-lactose) disaccharide was allo-lactose in both purified GOS preparations. The predominant trisaccharide in GOS-A and GOS-E was shown to be 3′-galactosyllactose, with lower levels of 6′-galactosyllactose and 4′-galactosyllactose. These three oligosaccharides have also been reported to occur in human milk. Purified GOS-A and GOS-E were shown to be able to support bifidobacterial growth similar to a commercially available GOS. In addition, GOS-E and the commercially available GOS were shown to be capable of reducing Escherichia coli adhesion to a C2BBe1 cell line. Both in vitro bifidogenic activity and reduced E. coli adhesion support the prebiotic potential of GOS-E and GOS-A.
Collapse
Affiliation(s)
- Valentina Ambrogi
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| | - Francesca Bottacini
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Biological Sciences, Munster Technological University, Cork, Ireland
| | - John Mac Sharry
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland.,School of Medicine, University College Cork, Cork, Ireland
| | | | - Ellen O'Keeffe
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Dan Walsh
- School of Microbiology, University College Cork, Cork, Ireland
| | | | - Linqiu Cao
- FrieslandCampina, Amersfoort, Netherlands
| | | | | | | | | | - Oswaldo Hernandez-Hernandez
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - F Javier Moreno
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Douwe van Sinderen
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
15
|
Khan I, Huang Z, Liang L, Li N, Ali Z, Ding L, Hong M, Shi H. Ammonia stress influences intestinal histomorphology, immune status and microbiota of Chinese striped-neck turtle (Mauremys sinensis). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 222:112471. [PMID: 34229168 DOI: 10.1016/j.ecoenv.2021.112471] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/22/2021] [Accepted: 06/26/2021] [Indexed: 06/13/2023]
Abstract
Ammonia is one of major pollutants in aquatic environment that induces severe stress and toxicity to organisms in aquatic system. The intestine acts a major defense line that protects living organisms from biotic and abiotic stresses. In the current study, we examined the effects of ammonia on intestinal histomorphology, transcriptional levels of intestinal barrier functioning genes and intestinal microbiota of Chinese striped-neck turtle (Mauremys sinensis). Thus, the turtles were placed in water with addition of ammonia at 0 (control), 100, 200 mg L-1 for 30 days. Our findings showed that ammonia reduced the villus length and induced the inflammatory cells appearance. In addition, the epithelial tight junction genes, claudin and zonola occludin significantly downregulated in ammonia exposed groups as compared to control group (P < 0.05). Similarly, the mRNA expression levels of MUC-2 gene also significantly decreased in ammonia treated groups (P < 0.05). However, the expression levels of intestinal immune related genes such as IL-10, IL-12, TGF-β1, TNF-α and IFN-γ significantly increased (P < 0.05). Furthermore, ammonia changed gut microbial diversity variedly. At the phylum levels, Firmicutes increased, whereas Bacteroidota, Desulfobacterota and Synergistota decreased significantly. Likewise, Lachnospiraceae, Bacteroides, Eubacteriaceae, Desulfovibrio, Muribaculaceae, Bilophila, Cloacibacillus, Christensenellaceae, Ruminococcus and Parabacteroides decreased while, Romboutsia and Turicibacter increased in ammonia exposed groups. In conclusion, ammonia at 100 and 200 mg L-1 could alter the intestinal barrier function and change the composition of intestinal microbiota, leading to bad health status in M. sinensis.
Collapse
Affiliation(s)
- Ijaz Khan
- Key Laboratory of Tropical Island Ecology, Ministry of Education, Hainan key Laboratory of Tropical Animal and Plant Ecology, College of Life Sciences, Hainan Normal University, Haikou 571158, China
| | - Zubin Huang
- Key Laboratory of Tropical Island Ecology, Ministry of Education, Hainan key Laboratory of Tropical Animal and Plant Ecology, College of Life Sciences, Hainan Normal University, Haikou 571158, China
| | - Liangyue Liang
- Key Laboratory of Tropical Island Ecology, Ministry of Education, Hainan key Laboratory of Tropical Animal and Plant Ecology, College of Life Sciences, Hainan Normal University, Haikou 571158, China
| | - Na Li
- Key Laboratory of Tropical Island Ecology, Ministry of Education, Hainan key Laboratory of Tropical Animal and Plant Ecology, College of Life Sciences, Hainan Normal University, Haikou 571158, China
| | - Zeeshan Ali
- Key Laboratory of Tropical Island Ecology, Ministry of Education, Hainan key Laboratory of Tropical Animal and Plant Ecology, College of Life Sciences, Hainan Normal University, Haikou 571158, China
| | - Li Ding
- Key Laboratory of Tropical Island Ecology, Ministry of Education, Hainan key Laboratory of Tropical Animal and Plant Ecology, College of Life Sciences, Hainan Normal University, Haikou 571158, China.
| | - Meiling Hong
- Key Laboratory of Tropical Island Ecology, Ministry of Education, Hainan key Laboratory of Tropical Animal and Plant Ecology, College of Life Sciences, Hainan Normal University, Haikou 571158, China.
| | - Haitao Shi
- Key Laboratory of Tropical Island Ecology, Ministry of Education, Hainan key Laboratory of Tropical Animal and Plant Ecology, College of Life Sciences, Hainan Normal University, Haikou 571158, China.
| |
Collapse
|
16
|
Figueroa-Lozano S, Akkerman R, Beukema M, van Leeuwen SS, Dijkhuizen L, de Vos P. 2′-Fucosyllactose impacts the expression of mucus-related genes in goblet cells and maintains barrier function of gut epithelial cells. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
17
|
Jakobsen LMA, Sundekilde UK, Andersen HJ, Kot W, Mejia JLC, Nielsen DS, Hansen AK, Bertram HC. Administration of Bovine Milk Oligosaccharide to Weaning Gnotobiotic Mice Inoculated with a Simplified Infant Type Microbiota. Microorganisms 2021; 9:microorganisms9051003. [PMID: 34066501 PMCID: PMC8148552 DOI: 10.3390/microorganisms9051003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/22/2021] [Accepted: 05/04/2021] [Indexed: 12/18/2022] Open
Abstract
Bovine milk oligosaccharides (BMO) share structural similarity to selected human milk oligosaccharides, which are natural prebiotics for infants. Thus, there is a potential in including BMOs as a prebiotic in infant formula. To examine the in vivo effect of BMO-supplementation on the infant gut microbiota, a BMO-rich diet (2% w/w) was fed to gnotobiotic mice (n = 11) inoculated with an infant type co-culture and compared with gnotobiotic mice receiving a control diet (n = 9). Nuclear magnetic resonance metabolomics in combination with high-throughput 16S rRNA gene amplicon sequencing was used to compare metabolic activity and microbiota composition in different compartments of the lower gastrointestinal tract. BMO components were detected in cecum and colon contents, revealing that BMO was available for the gut bacteria. The gut microbiota was dominated by Enterobacteriaceae and minor abundance of Lactobacilliaceae, while colonization of Bifidobacteriaceae did not succeed. Apart from a lower E. coli population in cecum content and lower formate (in colon) and succinate (in colon and cecum) concentrations, BMO supplementation did not result in significant changes in microbiota composition nor metabolic activity. The present study corroborates the importance of the presence of bifidobacteria for obtaining microbial-derived effects of milk oligosaccharides in the gastrointestinal tract.
Collapse
Affiliation(s)
- Louise Margrethe Arildsen Jakobsen
- Department of Food Science, Aarhus University, Agro Food Park 48, 8200 Aarhus N, Denmark; (U.K.S.); (H.C.B.)
- Correspondence: ; Tel.: +45-2073-1316
| | - Ulrik Kræmer Sundekilde
- Department of Food Science, Aarhus University, Agro Food Park 48, 8200 Aarhus N, Denmark; (U.K.S.); (H.C.B.)
| | | | - Witold Kot
- Department of Plant and Environmental Sciences, University of Copenhagen, Thorvaldsensvej 40, 1871 Frederiksberg C, Denmark;
| | - Josue Leonardo Castro Mejia
- Department of Food Science, University of Copenhagen, Rolighedsvej 30, 1958 Frederiksberg C, Denmark; (J.L.C.M.); (D.S.N.)
| | - Dennis Sandris Nielsen
- Department of Food Science, University of Copenhagen, Rolighedsvej 30, 1958 Frederiksberg C, Denmark; (J.L.C.M.); (D.S.N.)
| | - Axel Kornerup Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Grønnegårdsvej 17, 1870 Frederiksberg C, Denmark;
| | - Hanne Christine Bertram
- Department of Food Science, Aarhus University, Agro Food Park 48, 8200 Aarhus N, Denmark; (U.K.S.); (H.C.B.)
| |
Collapse
|
18
|
Hajjar R, Oliero M, Cuisiniere T, Fragoso G, Calvé A, Djediai S, Annabi B, Richard CS, Santos MM. Improvement of colonic healing and surgical recovery with perioperative supplementation of inulin and galacto-oligosaccharides. Clin Nutr 2021; 40:3842-3851. [PMID: 34130031 DOI: 10.1016/j.clnu.2021.04.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/08/2021] [Accepted: 04/19/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND AIMS Anastomotic leak (AL) is a major complication in colorectal surgery. Recent evidence suggests that the gut microbiota may affect healing and may cause or prevent AL. Butyrate is a beneficial short-chain fatty acid (SCFA) that is produced as a result of bacterial fermentation of dietary oligosaccharides and has been described as beneficial in the maintenance of colonic health. To assess the impact of oligosaccharides on colonic anastomotic healing in mice, we propose to modulate the microbiota with oligosaccharides to increase butyrate production via enhancement of butyrate-producing bacteria and, consequently, improve anastomotic healing in mice. METHODS Animal experiments were conducted in mice that were subjected to diets supplemented with inulin, galacto-oligosaccharides (GOS) or cellulose, as a control, for two weeks before undergoing a surgical colonic anastomosis. Macroscopic and histological assessment of the anastomosis was performed. Extent of epithelial proliferation was assessed by Ki-67 immunohistochemistry. Gelatin zymography was used to evaluate the extent of matrix metalloproteinase (MMP) hydrolytic activity. RESULTS Inulin and GOS diets were associated with increased butyrate production and better anastomotic healing. Histological analysis revealed an enhanced mucosal continuity, and this was associated with an increased re-epithelialization of the wound as determined by increased epithelial proliferation. Collagen concentration in peri-anastomotic tissue was higher with inulin and GOS diets and MMP activity, a marker of collagen degradation, was lower with both oligosaccharides. Inulin and GOS diets were further associated with lower bacterial translocation. CONCLUSIONS Dietary supplementation with inulin and GOS may improve anastomotic healing and reinforce the gut barrier in mice.
Collapse
Affiliation(s)
- Roy Hajjar
- Nutrition and Microbiome Laboratory, Institut du Cancer de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), 900 Rue Saint-Denis, Montréal, Québec, H2X 0A9, Canada; Digestive Surgery Service, Department of Surgery, Centre hospitalier de l'Université de Montréal (CHUM), 1000 Rue Saint-Denis, Montréal, Québec, H2X 0C1, Canada; Department of Surgery, Université de Montréal, 2900 Boulevard Edouard-Montpetit, Montréal, Québec, H3T 1J4, Canada
| | - Manon Oliero
- Nutrition and Microbiome Laboratory, Institut du Cancer de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), 900 Rue Saint-Denis, Montréal, Québec, H2X 0A9, Canada
| | - Thibault Cuisiniere
- Nutrition and Microbiome Laboratory, Institut du Cancer de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), 900 Rue Saint-Denis, Montréal, Québec, H2X 0A9, Canada
| | - Gabriela Fragoso
- Nutrition and Microbiome Laboratory, Institut du Cancer de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), 900 Rue Saint-Denis, Montréal, Québec, H2X 0A9, Canada
| | - Annie Calvé
- Nutrition and Microbiome Laboratory, Institut du Cancer de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), 900 Rue Saint-Denis, Montréal, Québec, H2X 0A9, Canada
| | - Souad Djediai
- Molecular Oncology Laboratory, Department of Chemistry, Université du Québec à Montréal (UQAM), P. O. Box 8888, Succ. Centre-Ville, Montréal, Québec, H3C 3P8, Canada
| | - Borhane Annabi
- Molecular Oncology Laboratory, Department of Chemistry, Université du Québec à Montréal (UQAM), P. O. Box 8888, Succ. Centre-Ville, Montréal, Québec, H3C 3P8, Canada
| | - Carole S Richard
- Digestive Surgery Service, Department of Surgery, Centre hospitalier de l'Université de Montréal (CHUM), 1000 Rue Saint-Denis, Montréal, Québec, H2X 0C1, Canada; Department of Surgery, Université de Montréal, 2900 Boulevard Edouard-Montpetit, Montréal, Québec, H3T 1J4, Canada
| | - Manuela M Santos
- Nutrition and Microbiome Laboratory, Institut du Cancer de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), 900 Rue Saint-Denis, Montréal, Québec, H2X 0A9, Canada; Department of Medicine, Université de Montréal, 2900 Boulevard Edouard-Montpetit, Montréal, Québec, H3T 1J4, Canada.
| |
Collapse
|
19
|
Catenza KF, Donkor KK. Recent approaches for the quantitative analysis of functional oligosaccharides used in the food industry: A review. Food Chem 2021; 355:129416. [PMID: 33774226 DOI: 10.1016/j.foodchem.2021.129416] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/15/2021] [Accepted: 02/17/2021] [Indexed: 02/07/2023]
Abstract
Functional oligosaccharides (OS) are diverse groups of carbohydrates that confer several health benefits stemming from their prebiotic activity. Commonly used oligosaccharides, fructooligosaccharides and galactooligosaccharides, are used in a wide range of applications from food ingredients to mimic the prebiotic activity of human milk oligosaccharides (HMOs) in infant formula to sugar and fat replacers in dairy and bakery products. However, while consumption of these compounds is associated with several positive health effects, increased consumption can cause intestinal discomfort and aggravation of intestinal bowel syndrome symptoms. Hence, it is essential to develop rapid and reliable techniques to quantify OS for quality control and proper assessment of their functionality in food and food products. The present review will focus on recent analytical techniques used to quantify OS in different matrices such as food and beverage products.
Collapse
Affiliation(s)
- K F Catenza
- Department of Physical Sciences (Chemistry), Thompson Rivers University, 805 TRU Way, Kamloops, BC V2C 0C8, Canada
| | - K K Donkor
- Department of Physical Sciences (Chemistry), Thompson Rivers University, 805 TRU Way, Kamloops, BC V2C 0C8, Canada.
| |
Collapse
|
20
|
Pothuraju R, Chaudhary S, Rachagani S, Kaur S, Roy HK, Bouvet M, Batra SK. Mucins, gut microbiota, and postbiotics role in colorectal cancer. Gut Microbes 2021; 13:1974795. [PMID: 34586012 PMCID: PMC8489937 DOI: 10.1080/19490976.2021.1974795] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/14/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023] Open
Abstract
An imbalance in the crosstalk between the host and gut microbiota affects the intestinal barrier function, which results in inflammatory diseases and colorectal cancer. The colon epithelium protects itself from a harsh environment and various pathogenic organisms by forming a double mucus layer, primarily comprising mucins. Recent studies are focusing on how dietary patterns alter the gut microbiota composition, which in turn regulates mucin expression and maintains the intestinal layers. In addition, modulation of gut microbiota by microbiotic therapy (involving fecal microbiota transplantation) has emerged as a significant factor in the pathologies associated with dysbiosis. Therefore, proper communication between host and gut microbiota via different dietary patterns (prebiotics and probiotics) is needed to maintain mucus composition, mucin synthesis, and regulation. Here, we review how the interactions between diet and gut microbiota and bacterial metabolites (postbiotics) regulate mucus layer functionalities and mucin expression in human health and disease.
Collapse
Affiliation(s)
- Ramesh Pothuraju
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sanjib Chaudhary
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Hemant K. Roy
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Michael Bouvet
- Division of Surgical Oncology, Department of Surgery, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
21
|
van Leeuwen SS, te Poele EM, Chatziioannou AC, Benjamins E, Haandrikman A, Dijkhuizen L. Goat Milk Oligosaccharides: Their Diversity, Quantity, and Functional Properties in Comparison to Human Milk Oligosaccharides. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:13469-13485. [PMID: 33141570 PMCID: PMC7705968 DOI: 10.1021/acs.jafc.0c03766] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Human milk is considered the golden standard in infant nutrition. Free oligosaccharides in human milk provide important health benefits. These oligosaccharides function as prebiotics, immune modulators, and pathogen inhibitors and were found to improve barrier function in the gut. Infant formulas nowadays often contain prebiotics but lack the specific functions of human milk oligosaccharides (hMOS). Milk from domesticated animals also contains milk oligosaccharides but at much lower levels and with less diversity. Goat milk contains significantly more oligosaccharides (gMOS) than bovine (bMOS) or sheep (sMOS) milk and also has a larger diversity of structures. This review summarizes structural studies, revealing a diversity of up to 77 annotated gMOS structures with almost 40 structures fully characterized. Quantitative studies of goat milk oligosaccharides range from 60 to 350 mg/L in mature milk and from 200 to 650 mg/L in colostrum. These levels are clearly lower than in human milk (5-20 g/L) but higher than in other domesticated dairy animals, e.g., bovine (30-60 mg/L) and sheep (20-40 mg/L). Finally, the review focuses on demonstrated and potential functionalities of gMOS. Some studies have shown anti-inflammatory effects of mixtures enriched in gMOS. Goat MOS also display prebiotic potential, particularly in stimulating growth of bifidobacteria preferentially. Although functional studies of gMOS are still limited, several structures are also found in human milk and have known functions as immune modulators and pathogen inhibitors. In conclusion, goat milk constitutes a promising alternative source for milk oligosaccharides, which can be used in infant formula.
Collapse
Affiliation(s)
- Sander S. van Leeuwen
- Department
of Laboratory Medicine, Cluster Human Nutrition and Health, University Medical Center Groningen (UMCG), Hanzeplein 1, 9713 GZ Groningen, Netherlands
- E-mail:
| | | | | | | | | | - Lubbert Dijkhuizen
- CarbExplore
Research BV, 9747 AN Groningen, Netherlands
- Department
of Microbial Physiology, Groningen Biomolecular Sciences and Biotechnology
Institute (GBB), University of Groningen, 9700 AB Groningen, Netherlands
| |
Collapse
|
22
|
Addai FP, Lin F, Wang T, Kosiba AA, Sheng P, Yu F, Gu J, Zhou Y, Shi H. Technical integrative approaches to cheese whey valorization towards sustainable environment. Food Funct 2020; 11:8407-8423. [PMID: 32955061 DOI: 10.1039/d0fo01484b] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Whey, a byproduct of cheese production, is often treated as an industrial dairy waste. A large volume of this product is disposed of annually due to inadequate bioconversion approaches. With its high pollutant load, disposal without pretreatment has raised a lot of environmental concerns alerting the need to seek optimal methods for adequately extracting and utilizing its organic content. In recent years, several techniques for whey valorization have emerged which may serve as interventionary measures against its environmental effects after disposal. In this review, we discuss five major approaches, by which whey can be converted into eco-friendly products, to significantly cut whey wastage. The approaches to whey valorization are therefore examined under the following perspectives: whey as a raw material for the production of bioethanol and prebiotic oligosaccharides via β-galactosidase and microbe catalyzed reactions, for the production of refined lactose as an excipient for pharmaceutical purposes, and the clinical significance of whey hydrolysates and their antifungal activity in food processing.
Collapse
Affiliation(s)
- Frank Peprah Addai
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Feng Lin
- Key Laboratory of Healthy Freshwater Aquaculture, Ministry of Agriculture, Zhejiang Institute of Freshwater Fisheries, Huzhou 313001, P. R. China
| | - Taotao Wang
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Anthony A Kosiba
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Pengcheng Sheng
- Key Laboratory of Healthy Freshwater Aquaculture, Ministry of Agriculture, Zhejiang Institute of Freshwater Fisheries, Huzhou 313001, P. R. China
| | - Feng Yu
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Jie Gu
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Yang Zhou
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Haifeng Shi
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| |
Collapse
|