1
|
Ren X, Cai S, Zhong Y, Tang L, Xiao M, Li S, Zhu C, Li D, Mou H, Fu X. Marine-Derived Fucose-Containing Carbohydrates: Review of Sources, Structure, and Beneficial Effects on Gastrointestinal Health. Foods 2024; 13:3460. [PMID: 39517244 PMCID: PMC11545675 DOI: 10.3390/foods13213460] [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: 09/30/2024] [Revised: 10/12/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Fucose, fucose-containing oligosaccharides, and fucose-containing polysaccharides have been widely applied in the fields of food and medicine, including applications in Helicobacter pylori eradication and renal function protection. Fucose-containing carbohydrates (FCCs) derived from marine organisms such as seaweed, invertebrates, microalgae, fungi, and bacteria have garnered growing attention due to their diverse bioactivities and potential therapeutic applications. Marine-derived FCCs characterized by high fucose residue content and extensive sulfate substitution, including fucoidan, fucosylated chondroitin sulfate, and fucose-rich microbial exopolysaccharides, have demonstrated significant potential in promoting gastrointestinal health. This review describes the unique structural features of FCCs and summarizes their health benefits, including regulation of gut microbiota, modulation of microbial metabolism, anti-adhesion activities against H. pylori and gut pathogens, protection against inflammatory injuries, and anti-tumor activities. Additionally, this review discusses the structural characteristics that influence the functional properties and the limitations related to the activity research and preparation processes of FCCs, providing a balanced perspective on the application potential and challenges of FCCs with specific structures for the regulation of gastrointestinal health and diseases.
Collapse
|
2
|
Huang H, Yu W, Xu X, Liu Y, Li J, Du G, Lv X, Liu L. Combinatorial Engineering of Escherichia coli for Enhancing 3-Fucosyllactose Production. ACS Synth Biol 2024; 13:1866-1878. [PMID: 38836566 DOI: 10.1021/acssynbio.4c00132] [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: 06/06/2024]
Abstract
3-Fucosyllactose (3-FL) is an important fucosylated human milk oligosaccharide (HMO) with biological functions such as promoting immunity and brain development. Therefore, the construction of microbial cell factories is a promising approach to synthesizing 3-FL from renewable feedstocks. In this study, a combinatorial engineering strategy was used to achieve efficient de novo 3-FL production in Escherichia coli. α-1,3-Fucosyltransferase (futM2) from Bacteroides gallinaceum was introduced into E. coli and optimized to create a 3-FL-producing chassis strain. Subsequently, the 3-FL titer increased to 5.2 g/L by improving the utilization of the precursor lactose and down-regulating the endogenous competitive pathways. Furthermore, a synthetic membraneless organelle system based on intrinsically disordered proteins was designed to spatially regulate the pathway enzymes, producing 7.3 g/L 3-FL. The supply of the cofactors NADPH and GTP was also enhanced, after which the 3-FL titer of engineered strain E26 was improved to 8.2 g/L in a shake flask and 10.8 g/L in a 3 L fermenter. In this study, we developed a valuable approach for constructing an efficient 3-FL-producing cell factory and provided a versatile workflow for other chassis cells and HMOs.
Collapse
Affiliation(s)
- Huiyuan Huang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Wenwen Yu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Xianhao Xu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Yanfeng Liu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Jianghua Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Guocheng Du
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Xueqin Lv
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Ministry of Education, Jiangnan University, Wuxi 214122, China
- Yixing Institute of Food Biotechnology Co., Ltd., Yixing 214200, China
| | - Long Liu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Ministry of Education, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
3
|
Cox AJ, Speer H, Radcliffe CR, Masocha K, Ramsey R, West NP, Pyne DB. Immunomodulatory effects of fucoidan in recreationally active adult males undertaking 3-weeks of intensified training. J Sports Sci 2023; 41:1875-1882. [PMID: 38247026 DOI: 10.1080/02640414.2024.2305007] [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: 07/21/2023] [Accepted: 01/03/2024] [Indexed: 01/23/2024]
Abstract
The aim of the current study was to determine whether daily fucoidan supplementation (Undaria pinnatifida extract containing >85% fucoidan, 1 g/day) for three-weeks in a double blind-placebo controlled cross-over trial (ACTRN12621000872831) could modulate alterations in faecal (calprotectin, lysozyme and IgA) and salivary (lactoferrin, lysozyme and IgA) markers of mucosal immune competence typically observed in response to both acute physical activity, and a period of intensified exercise training, in healthy recreationally active men (n = 12). Participants responded positively to the intensified training with 16-19% improvement in mean power that was not different between supplement groups. Faecal biomarkers and concentrations of lactoferrin, lysozyme and IgA from resting saliva samples were largely stable over the supplementation period. Concentrations of salivary biomarkers varied significantly over time in response to acute exercise, however differences between supplementation groups were modest. For salivary lysozyme, there was a trend for a lower magnitude of increase post-exercise (p = 0.08) and limited return towards pre-exercise in response to fucoidan. For salivary IgA, a greater acute exercise response was noted for IgA in response to fucoidan (~2.7-fold higher; p = 0.02). Different dosage and supplementation protocols and inclusion of additional immune markers should be considered in subsequent assessments of any potential benefits of fucoidan supplementation in healthy active adults.
Collapse
Affiliation(s)
- Amanda J Cox
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
| | - Hollie Speer
- Research Institute for Sport & Exercise, University of Canberra, Canberra, ACT, Australia
| | - Ceridwen R Radcliffe
- Research Institute for Sport & Exercise, University of Canberra, Canberra, ACT, Australia
| | - Kathleen Masocha
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Rebecca Ramsey
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Nicholas P West
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
| | - David B Pyne
- Research Institute for Sport & Exercise, University of Canberra, Canberra, ACT, Australia
| |
Collapse
|
4
|
Li C, Li M, Hu M, Zhang T. Metabolic Engineering of De Novo Pathway for the Production of 2'-Fucosyllactose in Escherichia coli. Mol Biotechnol 2023; 65:1485-1497. [PMID: 36652181 DOI: 10.1007/s12033-023-00657-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 01/05/2023] [Indexed: 01/19/2023]
Abstract
2'-Fucosyllactose (2'-FL), one of the most abundant oligosaccharides in human milk, has gained increased attention owing to its nutraceutical and pharmaceutical potential. However, limited availability and high-cost of preparation have limited its widespread application and in-depth investigation of its potential functions. Here, a modular pathway engineering was implemented to construct an Escherichia coli strain to improve the biosynthesis titer of 2'-FL. Before overexpression of manB, manC, gmd, wcaG, and heterologous expression of futC, genes wcaJ and lacZ encoding UDP-glucose lipid carrier transferase and β-galactosidase, respectively, were inactivated from E. coli BL21 (DE3) with the CRISPR-Cas9 system, which inhibited the production of 2'-FL. The results showed that final shake flask culture yielded a 3.8-fold increase in 2'-FL (0.98 g/L) from the engineered strain ELC07. Fed-batch fermentation conditions were optimized in a 3-L bioreactor. The highest titer of 2'-FL (18.22 g/L) was obtained, corresponding to a yield of 0.25 g/g glycerol and a substrate conversion of 0.88 g/g lactose.
Collapse
Affiliation(s)
- Chenchen Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Mengli Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Miaomiao Hu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Tao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China.
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
5
|
Asher AT, Mangel L, Ari JB, Gover O, Ahmad WA, Herzlich J, Mandel D, Schwartz B, Lubetzky R. Human Milk Oligosaccharide Profile across Lactation Stages in Israeli Women-A Prospective Observational Study. Nutrients 2023; 15:nu15112548. [PMID: 37299512 DOI: 10.3390/nu15112548] [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: 03/19/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Human milk oligosaccharides (HMOs) stimulate the growth of gut commensals, prevent the adhesion of enteropathogens and modulate host immunity. The major factors influencing variations in the HMO profile are polymorphisms in the secretor (Se) or Lewis (Le) gene, which affect the activity of the enzymes fucoslytransferase 2 and 3 (FUT2 and FUT3) that lead to the formation of four major fucosylated and non-fucosylated oligosaccharides (OS). This pilot study aimed to determine the HMO profile of Israeli breastfeeding mothers of 16 term and 4 preterm infants, from a single tertiary center in the Tel Aviv area. Fifty-two human milk samples were collected from 20 mothers at three-time points: colostrum, transitional milk and mature milk. The concentrations of nine HMOs were assessed using liquid chromatography coupled with mass spectra chromatograms. Fifty-five percent of the mothers were secretors and 45% were non-secretors. Infant sex affected HMO levels depending on the maternal secretor status. Secretor mothers to boys had higher levels of FUT2-dependent OS and higher levels of disialyllacto-N-tetraose in the milk of mothers to girls, whereas non-secretor mothers to girls had higher levels of 3'-sialyllactose. In addition, the season at which the human milk samples were obtained affected the levels of some HMOs, resulting in significantly lower levels in the summer. Our findings provide novel information on the irregularity in the HMO profile among Israeli lactating women and identify several factors contributing to this variability.
Collapse
Affiliation(s)
- Adi Talan Asher
- Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Laurence Mangel
- Tel Aviv Medical Center, Department of Neonatology, Dana Dwek Children's Hospital, Affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Julius Ben Ari
- The Interdepartmental Equipment Facility, Robert H. Smith Faculty of Agriculture, Food and Environment, Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Ofer Gover
- Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Wiessam Abu Ahmad
- School of Public Health and Community Medicine, The Hebrew University of Jerusalem, Ein Kerem. P.O. Box 12271, Jerusalem 9112102, Israel
| | - Jacky Herzlich
- Tel Aviv Medical Center, Department of Neonatology, Dana Dwek Children's Hospital, Affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dror Mandel
- Tel Aviv Medical Center, Department of Neonatology, Dana Dwek Children's Hospital, Affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Betty Schwartz
- Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Ronit Lubetzky
- Tel Aviv Medical Center, Department of Neonatology, Dana Dwek Children's Hospital, Affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
6
|
Kaplan M, Şahutoğlu AS, Sarıtaş S, Duman H, Arslan A, Pekdemir B, Karav S. Role of milk glycome in prevention, treatment, and recovery of COVID-19. Front Nutr 2022; 9:1033779. [PMID: 36424926 PMCID: PMC9680090 DOI: 10.3389/fnut.2022.1033779] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/10/2022] [Indexed: 08/23/2023] Open
Abstract
Milk contains all essential macro and micro-nutrients for the development of the newborn. Its high therapeutic and antimicrobial content provides an important function for the prevention, treatment, and recovery of certain diseases throughout life. The bioactive components found in milk are mostly decorated with glycans, which provide proper formation and modulate the biological functions of glycosylated compounds. The glycome of milk consists of free glycans, glycolipids, and N- and O- glycosylated proteins. Recent studies have shown that both free glycans and glycan-containing molecules have antiviral characteristics based on different mechanisms such as signaling, microbiome modulation, natural decoy strategy, and immunomodulatory action. In this review, we discuss the recent clinical studies and potential mechanisms of free and conjugated glycans' role in the prevention, treatment, and recovery of COVID-19.
Collapse
Affiliation(s)
- Merve Kaplan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | | | - Sümeyye Sarıtaş
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Ayşenur Arslan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Burcu Pekdemir
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| |
Collapse
|
7
|
Eun Pak M, Kim YJ, Jin Park Y, Go Y, Soo Shin C, Yoon JW, Jeon SM, Song YH, Kim K. Human milk oligosaccharide, 2′-Fucosyllactose, attenuates platelet activation in arterial thrombosis. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
8
|
Phipps KR, Lozon D, Stannard DR, Gilby B, Baldwin N, Mikš MH, Lau A, Röhrig CH. Neonatal subchronic toxicity and in vitro genotoxicity studies of the human-identical milk oligosaccharide 3-fucosyllactose. J Appl Toxicol 2022; 42:1671-1687. [PMID: 35510931 DOI: 10.1002/jat.4335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/01/2022] [Indexed: 11/10/2022]
Abstract
Human milk oligosaccharides, such as 3-fucosyllactose (3-FL), are bioactive components of breast milk associated with benefits for infant growth and development. Structurally identical compounds (human-identical milk oligosaccharides - HiMOs) can be produced using microbial fermentation, allowing their use in infant formula to increase its similarity with human milk. Toxicological studies are required to demonstrate safety of HiMOs and that of any impurities potentially carried over from the manufacturing process. Biotechnologically produced 3-FL was tested for potential genotoxicity (bacterial reverse mutation test and in vitro mammalian micronucleus test) and subchronic toxicity (90-day study with neonatal rats). In the 90-day study, 3-FL was administered by gavage to rats once daily from Day 7 of age, at doses up to 4000 mg/kg body weight (bw)/day (the maximum feasible dose), followed by a 4-week recovery period. Reference controls received 4000 mg/kg bw/day of oligofructose, an ingredient permitted for use in infant formula. Results for the genotoxicity studies were negative. In the 90-day study, there were no adverse effects of 3-FL on any of the parameters measured; thus, the no-observed-adverse-effect level was 4000 mg/kg bw/day (the highest dose tested). These results support the safety of biotechnologically produced 3-FL for use in infant formula and other foods.
Collapse
Affiliation(s)
| | - Dayna Lozon
- Intertek Health Sciences Inc., Mississauga, Ontario, Canada
| | - Diane R Stannard
- Labcorp Early Development Laboratories Limited (formerly Covance Laboratories Limited), Eye, Suffolk, UK
| | - Ben Gilby
- Labcorp Early Development Laboratories Limited (formerly Covance Laboratories Limited), Woolley Road, Alconbury, Huntingdon, Cambridgeshire, UK
| | | | - Marta Hanna Mikš
- Glycom A/S, Hørsholm, Denmark.,University of Warmia and Mazury in Olsztyn, Faculty of Food Science, Olsztyn, Poland
| | | | | |
Collapse
|
9
|
Li Z, Zhu Y, Ni D, Zhang W, Mu W. Occurrence, functional properties, and preparation of 3-fucosyllactose, one of the smallest human milk oligosaccharides. Crit Rev Food Sci Nutr 2022; 63:9364-9378. [PMID: 35438024 DOI: 10.1080/10408398.2022.2064813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Human milk oligosaccharides (HMOs) are receiving wide interest and high attention due to their health benefits, especially for newborns. The HMOs-fortified products are expected to mimic human milk not only in the kinds of added oligosaccharides components but also the appropriate proportion between these components, and further provide the nutrition and physiological effects of human milk to newborns as closely as possible. In comparison to intensively studied 2'-fucosyllactose (2'-FL), 3-fucosyllactose (3-FL) has less attention in almost all respects. Nerveless, 3-FL naturally occurs in breast milk and increases roughly over the course of lactation with a nonnegligible content, and plays an irreplaceable role in human milk and delivers functional properties to newborns. According to the safety evaluation, 3-FL shows no acute oral toxicity, genetic toxicity, and subchronic toxicity. It has been approved as generally recognized as safe (GRAS). Biological production of 3-FL can be realized by enzymatic and cell factory approaches. The α1,3- or α1,3/4-fucosyltransferase is the key enzyme for 3-FL biosynthesis. Various metabolic engineering strategies have been applied to enhance 3-FL yield using cell factory approach. In conclusion, this review gives an overview of the recent scientific literatures regarding occurrence, bioactive properties, safety evaluation, and biotechnological preparation of 3-FL.
Collapse
Affiliation(s)
- Zeyu Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Dawei Ni
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wenli Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| |
Collapse
|
10
|
Sprenger N, Tytgat HL, Binia A, Austin S, Singhal A. Biology of human milk oligosaccharides: from Basic Science to Clinical Evidence. J Hum Nutr Diet 2022; 35:280-299. [PMID: 35040200 PMCID: PMC9304252 DOI: 10.1111/jhn.12990] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/05/2022] [Indexed: 11/28/2022]
Abstract
Human milk oligosaccharides (HMOs) have been researched by scientists for over 100 years, driven by the substantial evidence for the nutritional and health benefits of mother's milk. Yet research has truly bloomed during the last decade, thanks to the progress in biotechnology, which allowed the production of large amounts of bona fide HMOs. The availability of HMOs has been particularly crucial for the renewed interest in HMO research because of the low abundance or even absence of HMOs in farmed animal milk. This interest is reflected in the increasing number of original research publications and reviews on HMOs. Here, we provide an overview and critical discussion on structure function relations of HMOs that highlight why they are such interesting and important components of human milk. Clinical observations in breastfed infants backed by basic research from animal models provide guidance as to what physiological roles for HMOs are to be expected. From an evidence-based nutrition viewpoint, we discuss the current data supporting clinical relevance of specific HMOs based on randomized placebo controlled clinical intervention trials in formula-fed infants. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Norbert Sprenger
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Hanne Lp Tytgat
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Aristea Binia
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Sean Austin
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Atul Singhal
- Institute of Child Health, University College London, London, WC1N 1EH, United Kingdom
| |
Collapse
|
11
|
Six Oligosaccharides' Variation in Breast Milk: A Study in South China from 0 to 400 Days Postpartum. Nutrients 2021; 13:nu13114017. [PMID: 34836272 PMCID: PMC8623037 DOI: 10.3390/nu13114017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/31/2021] [Accepted: 11/05/2021] [Indexed: 12/15/2022] Open
Abstract
This study investigated the variation in oligosaccharide levels in the breast milk of south Chinese mothers in a prolonged breastfeeding period of up to 400 days postpartum. A total of 488 breast milk samples were collected from 335 healthy mothers at five different time points: 0–5 days, 10–15 days, 40–45 days, 200–240 days, and 300–400 days postpartum. A high-performance anion-exchange chromatography-pulsed amperometric detector (HPAEC-PAD) was used to quantify 2′-fucosyllactose (2′-FL), 3-fucosyllactose (3-FL), lacto-N-tetraose (LNT), lacto-N-neotetraose (LNnT), 3′-sialyllactose (3′-SL) and 6′-sialyllactose (6′-SL). In this study, we found six oligosaccharides that were present in breast milk from 0 to 400 days postpartum. The median value ranges of individual oligosaccharide components in this study were 1013–2891 mg/L 2′-FL, 193–1421 mg/L 3-FL, 314–1478 mg/L LNT, 44–255 mg/L LNnT, 111–241 mg/L 3′-SL, and 23–602 mg/L6′-SL. HMO levels decreased over the lactation periods, except for 3-FL, which increased throughout lactation. The predominant fucosylated and sialylated HMOs were 2′-FL and 6′-SL at 40–45 days postpartum and changed to 3-FL and 3′-SL at 200–240 days postpartum. Results from this study showed that lactating women continue to provide their offspring with a high level of 2′-FL one year after delivery, suggesting that 2′-FL may play an important role for infants in early life. Our findings also provide further evidence in support of breastfeeding after one-year postpartum.
Collapse
|
12
|
Abstract
The neonatal body provides a range of potential habitats, such as the gut, for microbes. These sites eventually harbor microbial communities (microbiotas). A "complete" (adult) gut microbiota is not acquired by the neonate immediately after birth. Rather, the exclusive, milk-based nutrition of the infant encourages the assemblage of a gut microbiota of low diversity, usually dominated by bifidobacterial species. The maternal fecal microbiota is an important source of bacterial species that colonize the gut of infants, at least in the short-term. However, development of the microbiota is influenced by the use of human milk (breast feeding), infant formula, preterm delivery of infants, caesarean delivery, antibiotic administration, family details and other environmental factors. Following the introduction of weaning (complementary) foods, the gut microbiota develops in complexity due to the availability of a diversity of plant glycans in fruits and vegetables. These glycans provide growth substrates for the bacterial families (such as members of the Ruminococcaceae and Lachnospiraceae) that, in due course, will dominate the gut microbiota of the adult. Although current data are often fragmentary and observational, it can be concluded that the nutrition that a child receives in early life is likely to impinge not only on the development of the microbiota at that time but also on the subsequent lifelong, functional relationships between the microbiota and the human host. The purpose of this review, therefore, is to discuss the importance of promoting the assemblage of functionally robust gut microbiotas at appropriate times in early life.
Collapse
Affiliation(s)
- Gerald W. Tannock
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
13
|
Ferrier L, Eutamène H, Siegwald L, Marquard AM, Tondereau V, Chevalier J, Jacot GE, Favre L, Theodorou V, Vicario M, Rytz A, Bergonzelli G, Garcia-Rodenas CL. Human milk oligosaccharides alleviate stress-induced visceral hypersensitivity and associated microbiota dysbiosis. J Nutr Biochem 2021; 99:108865. [PMID: 34582967 DOI: 10.1016/j.jnutbio.2021.108865] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 06/11/2021] [Accepted: 09/01/2021] [Indexed: 02/08/2023]
Abstract
Pain-related functional gastrointestinal disorders (FGIDs) are characterized by visceral hypersensitivity (VHS) associated with alterations in the microbiota-gut-brain axis. Since human milk oligosaccharides (HMOs) modulate microbiota, gut and brain, we investigated whether HMOs impact VHS, and explored the role of gut microbiota. To induce VHS, C57BL/6JRj mice received hourly water avoidance stress (WAS) sessions for 10 d, or antibiotics (ATB) for 12 d. Challenged and unchallenged (Sham) animals were fed AIN93M diet (Cont) or AIN93M containing 1% of a 6-HMO mix (HMO6). VHS was assessed by monitoring the visceromotor response to colorectal distension. Fecal microbiome was analyzed by shotgun metagenomics. The effect of HMO6 sub-blends on VHS and nociceptive pathways was further tested using the WAS model. In mice fed Cont, WAS and ATB increased the visceromotor response to distension. HMO6 decreased WAS-mediated electromyographic rise at most distension volumes and overall Area Under Curve (AUC=6.12±0.50 in WAS/HMO6 vs. 9.46±0.50 in WAS/Cont; P<.0001). In contrast, VHS in ATB animals was not improved by HMO6. In WAS, HMO6 promoted most microbiota taxa and several functional pathways associated with low VHS and decreased those associated with high VHS. Among the sub-blends, 2'FL+DFL and LNT+6'SL reduced visceromotor response close to Sham/Cont values and modulated serotoninergic and CGRPα-related pathways. This research further substantiates the capacity of HMOs to modulate the microbiota-gut-brain communication and identifies mitigation of abdominal pain as a new HMO benefit. Ultimately, our findings suggest the value of specific HMO blends to alleviate pain associated FGIDs such as infantile colic or Irritable Bowel Syndrome.
Collapse
Affiliation(s)
- Laurent Ferrier
- Nestlé Institute of Health Sciences, Nestle Research, Lausanne, Switzerland
| | - Hélène Eutamène
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Léa Siegwald
- Nestlé Institute of Health Sciences, Nestle Research, Lausanne, Switzerland
| | | | - Valerie Tondereau
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Julien Chevalier
- Nestlé Institute of Health Sciences, Nestle Research, Lausanne, Switzerland
| | - Guillaume E Jacot
- Nestlé Institute of Health Sciences, Nestle Research, Lausanne, Switzerland
| | - Laurent Favre
- Project Management, Nestle Research, Lausanne, Switzerland
| | - Vassilia Theodorou
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Maria Vicario
- Nestlé Institute of Health Sciences, Nestle Research, Lausanne, Switzerland
| | - Andreas Rytz
- Clinical Research Unit, Nestle Research, Lausanne, Switzerland
| | | | | |
Collapse
|
14
|
Hill DR, Chow JM, Buck RH. Multifunctional Benefits of Prevalent HMOs: Implications for Infant Health. Nutrients 2021; 13:3364. [PMID: 34684364 PMCID: PMC8539508 DOI: 10.3390/nu13103364] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 12/13/2022] Open
Abstract
Breastfeeding is the best source of nutrition during infancy and is associated with a broad range of health benefits. However, there remains a significant and persistent need for innovations in infant formula that will allow infants to access a wider spectrum of benefits available to breastfed infants. The addition of human milk oligosaccharides (HMOs) to infant formulas represents the most significant innovation in infant nutrition in recent years. Although not a direct source of calories in milk, HMOs serve as potent prebiotics, versatile anti-infective agents, and key support for neurocognitive development. Continuing improvements in food science will facilitate production of a wide range of HMO structures in the years to come. In this review, we evaluate the relationship between HMO structure and functional benefits. We propose that infant formula fortification strategies should aim to recapitulate a broad range of benefits to support digestive health, immunity, and cognitive development associated with HMOs in breastmilk. We conclude that acetylated, fucosylated, and sialylated HMOs likely confer important health benefits through multiple complementary mechanisms of action.
Collapse
Affiliation(s)
| | | | - Rachael H. Buck
- Abbott Nutrition, 3300 Stelzer Road, Columbus, OH 43219, USA; (D.R.H.); (J.M.C.)
| |
Collapse
|
15
|
Han SM, Derraik JGB, Binia A, Sprenger N, Vickers MH, Cutfield WS. Maternal and Infant Factors Influencing Human Milk Oligosaccharide Composition: Beyond Maternal Genetics. J Nutr 2021; 151:1383-1393. [PMID: 33768224 DOI: 10.1093/jn/nxab028] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 01/06/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Maternal genetics is a key determinant of human milk oligosaccharide (HMO) composition in human milk. Beyond genetic status, other factors influencing the HMO profile are poorly defined. Thus, we aimed to review the existing evidence on the associations between nongenetic maternal and infant factors and HMO composition. A systematic search was performed on PubMed and Web of Science (without a time restriction) to identify any relevant studies published. In total, 1056 results were obtained, of which 29 articles were selected to be included in this review. The range of factors investigated include lactation stage, maternal pre-pregnancy BMI (ppBMI), maternal age, parity, maternal diet, mode of delivery, infant gestational age, and infant sex. The data suggest that, beyond maternal genetics, HMO composition seems to be influenced by all these factors, but the underlining mechanisms remain speculative. The published evidence is discussed in this review, along with potential implications for infant growth and development. For example, 2'-fucosyllactose, which was reportedly increased in mothers with higher ppBMIs, was also associated with increased infant weight and height. In addition, greater levels of sialylated HMOs after preterm birth may support brain development in these infants.
Collapse
Affiliation(s)
- Soo Min Han
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - José G B Derraik
- Liggins Institute, The University of Auckland, Auckland, New Zealand.,Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden.,Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Aristea Binia
- Nestlé Research, Société des Produits Nestlé SA, Lausanne, Switzerland
| | - Norbert Sprenger
- Nestlé Research, Société des Produits Nestlé SA, Lausanne, Switzerland
| | - Mark H Vickers
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Wayne S Cutfield
- Liggins Institute, The University of Auckland, Auckland, New Zealand.,A Better Start-National Science Challenge, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
16
|
Morrin ST, Buck RH, Farrow M, Hickey RM. Milk-derived anti-infectives and their potential to combat bacterial and viral infection. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104442] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
17
|
Duman H, Kaplan M, Arslan A, Sahutoglu AS, Kayili HM, Frese SA, Karav S. Potential Applications of Endo-β- N-Acetylglucosaminidases From Bifidobacterium longum Subspecies infantis in Designing Value-Added, Next-Generation Infant Formulas. Front Nutr 2021; 8:646275. [PMID: 33898500 PMCID: PMC8063050 DOI: 10.3389/fnut.2021.646275] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Human milk is the optimal source of infant nutrition. Among many other health benefits, human milk can stimulate the development of a Bifidobacterium-rich microbiome through human milk oligosaccharides (HMOs). In recent years, the development of novel formulas has placed particular focus on incorporating some of the beneficial functional properties of human milk. These include adding specific glycans aimed to selectively stimulate the growth of Bifidobacterium. However, the bifidogenicity of human milk remains unparalleled. Dietary N-glycans are carbohydrate structures conjugated to a wide variety of glycoproteins. These glycans have a remarkable structural similarity to HMOs and, when released, show a strong bifidogenic effect. This review discusses the biocatalytic potential of the endo-β-N-acetylglucosaminidase enzyme (EndoBI-1) from Bifidobacterium longum subspecies infantis (B. infantis), in releasing N-glycans inherently present in infant formula as means to increase the bifidogenicity of infant formula. Finally, the potential implications for protein deglycosylation with EndoBI-1 in the development of value added, next-generation formulas are discussed from a technical perspective.
Collapse
Affiliation(s)
- Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Merve Kaplan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Ayşenur Arslan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | | | - Haci Mehmet Kayili
- Department of Biomedical Engineering, Karabuk University, Karabük, Turkey
| | - Steven A Frese
- Department of Nutrition, University of Nevada, Reno, NV, United States.,Department of Food Science and Technology, University of Nebraska Lincoln, Lincoln, NE, United States
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| |
Collapse
|
18
|
Oba PM, Lee AH, Vidal S, Wyss R, Miao Y, Adesokan Y, Swanson KS. Effect of a novel animal milk oligosaccharide biosimilar on macronutrient digestibility and gastrointestinal tolerance, fecal metabolites, and fecal microbiota of healthy adult cats. J Anim Sci 2021; 99:skaa399. [PMID: 33320182 PMCID: PMC7799586 DOI: 10.1093/jas/skaa399] [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: 09/18/2020] [Accepted: 12/11/2020] [Indexed: 11/13/2022] Open
Abstract
GNU100 is a novel animal milk oligosaccharide (AMO) biosimilar. In a recent in vitro fermentation study, GNU100 was shown to be fermentable by feline gastrointestinal microbiota and lead to increased short-chain fatty acid production. Our objectives herein were to evaluate the palatability, safety, and gastrointestinal tolerance of GNU100 in healthy adult cats. Exploratory end-points were measured to assess utility. In study 1, 20 adult cats were used to test the palatability of diets containing 0% or 1% GNU100. In study 2, 32 (mean age = 1.9 yr; mean body weight = 4.6 kg) male (n = 12) and female (n = 20) adult cats were used in a completely randomized design. After a 2-wk baseline, cats were assigned to one of the following treatment groups and fed for 26 wk: control (CT, no GNU100), low dose (LD, 0.5% GNU100), medium dose (MD, 1.0% GNU100), and high dose (HD, 1.5% GNU100). On weeks 2, 4, and 26, fresh fecal samples were collected for the measurement of stool quality and immune and inflammatory markers and on weeks 2 and 4 for microbiota and metabolites. On week 4, total feces were collected to measure apparent total tract macronutrient digestibility. On weeks 2, 4, and 26, blood samples were collected for serum chemistry, hematology, and inflammatory marker measurement. The palatability test showed that 1% GNU100 was strongly preferred (P < 0.05), with GNU100 having a 17.6:1 consumption ratio compared with control. In the long-term study, all cats remained healthy, without any signs of gastrointestinal intolerance or illness. All diets were well accepted, resulting in similar (P > 0.05) food intake, fecal characteristics, immunoglobulin A, and calprotectin, and dry matter, organic matter, fat, and crude protein digestibilities. Fecal butyrate was greater (P = 0.02) in cats fed HD than cats fed LD or MD. Fecal indole was lower (P = 0.02) in cats fed HD than cats fed LD. Cats fed CT had a higher (P = 0.003) relative abundance of Actinobacteria than cats fed LD. The relative abundance of Peptococcus was impacted by diet and time. At 4 wk, Campylobacter was lower in fecal samples of cats fed HD. Overall, the data suggest that dietary GNU100 supplementation was highly palatable, well tolerated, did not cause detrimental effects on fecal quality or nutrient digestibility, increased fecal butyrate concentrations, and reduced fecal indole concentrations, supporting the safety of GNU100 for inclusion in feline diets and suggesting potential benefits on gastrointestinal health of cats.
Collapse
Affiliation(s)
- Patrícia M Oba
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Anne H Lee
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Sara Vidal
- Gnubiotics Sciences SA, Epalinges, Switzerland
| | - Romain Wyss
- Gnubiotics Sciences SA, Epalinges, Switzerland
| | - Yong Miao
- Gnubiotics Sciences SA, Epalinges, Switzerland
| | | | - Kelly S Swanson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
| |
Collapse
|
19
|
Oba PM, Vidal S, Wyss R, Miao Y, Adesokan Y, Swanson KS. Effect of a novel animal milk oligosaccharide biosimilar on the gut microbial communities and metabolites of in vitro incubations using feline and canine fecal inocula. J Anim Sci 2020; 98:5897395. [PMID: 32845316 DOI: 10.1093/jas/skaa273] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/20/2020] [Indexed: 12/17/2022] Open
Abstract
Milk oligosaccharides (MO) confer multiple potential physiological benefits, such as the selective growth promotion of beneficial microbiota, inhibition of enteric pathogen growth and adhesion to enterocytes, maturation of the gut mucosal barrier, and modulation of the gastrointestinal immune system. This study was conducted to determine the fermentation potential of GNU100, an animal MO biosimilar, in an in vitro system using healthy canine and feline fecal inocula. Single feline and single canine fecal samples were used to inoculate a batch fermentation system. Tubes containing a blank control (BNC), GNU100 at 0.5% (5 g/L; GNU1), or GNU100 at 1.0% (10 g/L; GNU2) were incubated for 48 h. Gas pressure, pH, lactate, short-chain fatty acids (SCFA; acetate, propionate, and butyrate), and branched-chain fatty acids (BCFA; isobutyrate, isovalerate, and valerate) were measured after 6, 24, and 48 h. Ammonium and microbiota (total bacteria by flow cytometry and Pet-16Seq; Lactobacillus and Bifidobacterium by quantitative polymerase chain reaction ) were measured after 24 and 48 h. Data were analyzed using the Mixed Models procedure of SAS. Substrates were considered to be a fixed effect and replicates considered to be a random effect. Tukey's multiple comparison analysis was used to compare least squares means, with differences considered significant with P < 0.05. In feline and canine incubations, SCFA increases were greater (P < 0.0001) in GNU100 compared with BNC, with acetate making up the largest SCFA proportion (P < 0.0001). GNU100 cultures led to greater increases (P < 0.0001) in lactate and ammonium than BNC in the feline incubations. GNU100 cultures led to greater increases (P < 0.0001) in ammonium than BNC in canine incubations and greater increases (P < 0.0001) in BCFA than BNC in feline incubations. Pet-16Seq microbial profiles from the feline and canine fecal incubations exhibited a modulation after GNU100 fermentation, with a reduction of the genera Escherichia/Shigella and Salmonella. In feline incubations, Bifidobacterium populations had greater increases (P < 0.0001) in GNU100 than BNC. In feline incubations, Lactobacillus populations had greater increases (P = 0.01) in GNU100 than BNC, with GNU1 leading to greater increases (P = 0.02) in Lactobacillus than BNC tubes in canine incubations. Overall, this study demonstrated that GNU100 was fermented in an in vitro fermentation system inoculated with canine and feline microbiota, resulting in the growth of beneficial bacteria and the production of SCFA, BCFA, and ammonium.
Collapse
Affiliation(s)
- Patrícia M Oba
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Sara Vidal
- Gnubiotics Sciences SA, Épalinges, Switzerland
| | - Romain Wyss
- Gnubiotics Sciences SA, Épalinges, Switzerland
| | - Yong Miao
- Gnubiotics Sciences SA, Épalinges, Switzerland
| | | | - Kelly S Swanson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL.,Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
| |
Collapse
|
20
|
Abstract
Numerous bioactive components exist in human milk including free oligosaccharides, which represent some of the most important, and provide numerous health benefits to the neonate. Considering the demonstrated value of these compounds, much interest lies in characterising structurally similar oligosaccharides in the dairy industry. In this study, the impacts of days post-parturition and parity of the cows on the oligosaccharide and lactose profiles of their milk were evaluated. Colostrum and milk samples were obtained from 18 cows 1–5 days after parturition. Three distinct phases were identified using multivariate analysis: colostrum (day 0), transitional milk (days 1–2) and mature milk (days 3–5). LS-tetrasaccharide c, lacto-N-neotetraose, disialyllacto-N-tetraose, 3’-sial-N-acetyllactosamine, 3’-sialyllactose, lacto-N-neohexaose and disialyllactose were found to be highly affiliated with colostrum. Notably, levels of lactose were at their lowest concentration in the colostrum and substantially increased 1-day post-parturition. The cow’s parity was also shown to have a significant effect on the oligosaccharide profile, with first lactation cows containing more disialyllacto-N-tetraose, 6’-sialyllactose and LS-tetrasaccharide compared to cows in their second or third parity. Overall, this study identifies key changes in oligosaccharide and lactose content that clearly distinguish colostrum from transitional and mature milk and may facilitate the collection of specific streams with divergent biological functions.
Collapse
|
21
|
Palsson OS, Peery A, Seitzberg D, Amundsen ID, McConnell B, Simrén M. Human Milk Oligosaccharides Support Normal Bowel Function and Improve Symptoms of Irritable Bowel Syndrome: A Multicenter, Open-Label Trial. Clin Transl Gastroenterol 2020; 11:e00276. [PMID: 33512807 PMCID: PMC7721220 DOI: 10.14309/ctg.0000000000000276] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/21/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Treatment options for irritable bowel syndrome (IBS) are limited, causing many patients to remain symptomatic. This study assessed the potential of human milk oligosaccharides (HMOs) to normalize bowel habits. Secondary outcomes included IBS severity and health-related quality of life. METHODS This multicenter, open-label trial recruited patients with IBS from 17 sites across the United States. Patients received daily orally administrated 5-g intervention of the HMOs 2'-fucosyllactose and lacto-N-neotetraose in a 4:1 mix. Bowel habits, IBS symptoms, and quality of life were assessed at baseline and every 4 weeks during the 12-week intervention. RESULTS A total of 317 patients (70.7% women; mean age of 44.0 years, range 18-93 years) received the trial product, and 245 patients completed the trial according to protocol. Patients had a significant improvement from baseline to 12 weeks in total percentage of bowel movements with abnormal stool consistency (mean and [95% confidence interval]: 90.7 [88.9-92.9] vs 57.2% [53.9-60.5], P < 0.0001), overall IBS Symptom Severity Score (323 [314-332] vs 144 [133-155], P < 0.0001) and health-rela,ted quality of life (50.4 [48.0-52.8] vs 74.6 [72.3-76.9], P < 0.0001). Improvement was similar across IBS subtypes. Symptoms improved most in the first 4 weeks of intervention. The most common side effects were mild gastrointestinal symptoms such as flatulence, abdominal pain and discomfort, and distension. DISCUSSION Supplementation with 2 selected HMOs improves IBS symptoms and quality of life without substantial side effects. These promising results suggest that this novel approach to IBS should be confirmed in a randomized, placebo-controlled trial.
Collapse
Affiliation(s)
- Olafur S. Palsson
- Center for Functional Gastrointestinal and Motility Disorders, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Anne Peery
- Center for Functional Gastrointestinal and Motility Disorders, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | | | | | - Magnus Simrén
- Center for Functional Gastrointestinal and Motility Disorders, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
22
|
In Love with Shaping You-Influential Factors on the Breast Milk Content of Human Milk Oligosaccharides and Their Decisive Roles for Neonatal Development. Nutrients 2020; 12:nu12113568. [PMID: 33233832 PMCID: PMC7699834 DOI: 10.3390/nu12113568] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/08/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023] Open
Abstract
Human milk oligosaccharides (HMOs) are structurally versatile sugar molecules constituting the third major group of soluble components in human breast milk. Based on the disaccharide lactose, the mammary glands of future and lactating mothers produce a few hundreds of different HMOs implicating that their overall anabolism utilizes rather high amounts of energy. At first sight, it therefore seems contradictory that these sugars are indigestible for infants raising the question of why such an energy-intensive molecular class evolved. However, in-depth analysis of their molecular modes of action reveals that Mother Nature created HMOs for neonatal development, protection and promotion of health. This is not solely facilitated by HMOs in their indigestible form but also by catabolites that are generated by microbial metabolism in the neonatal gut additionally qualifying HMOs as natural prebiotics. This narrative review elucidates factors influencing the HMO composition as well as physiological roles of HMOs on their way through the infant body and within the gut, where a major portion of HMOs faces microbial catabolism. Concurrently, this work summarizes in vitro, preclinical and observational as well as interventional clinical studies that analyzed potential health effects that have been demonstrated by or were related to either human milk-derived or synthetic HMOs or HMO fractions.
Collapse
|
23
|
Al-Khafaji AH, Jepsen SD, Christensen KR, Vigsnæs LK. The potential of human milk oligosaccharides to impact the microbiota-gut-brain axis through modulation of the gut microbiota. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104176] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
24
|
Pérez-Escalante E, Alatorre-Santamaría S, Castañeda-Ovando A, Salazar-Pereda V, Bautista-Ávila M, Cruz-Guerrero AE, Flores-Aguilar JF, González-Olivares LG. Human milk oligosaccharides as bioactive compounds in infant formula: recent advances and trends in synthetic methods. Crit Rev Food Sci Nutr 2020; 62:181-214. [DOI: 10.1080/10408398.2020.1813683] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Emmanuel Pérez-Escalante
- Universidad Autónoma del Estado de Hidalgo, Área Académica de Química. Ciudad del Conocimiento, Carretera Pachuca-Tulancingo km 4.5, Colonia Carboneras. CP. 42184. Mineral de la Reforma, Hidalgo, México
| | - Sergio Alatorre-Santamaría
- Universidad Autónoma Metropolitana, Unidad Iztapalapa. División de Ciencias Biológicas y de la Salud. Departamento de Biotecnología, Colonia Vicentina AP 09340, Ciudad de México, México
| | - Araceli Castañeda-Ovando
- Universidad Autónoma del Estado de Hidalgo, Área Académica de Química. Ciudad del Conocimiento, Carretera Pachuca-Tulancingo km 4.5, Colonia Carboneras. CP. 42184. Mineral de la Reforma, Hidalgo, México
| | - Verónica Salazar-Pereda
- Universidad Autónoma del Estado de Hidalgo, Área Académica de Química. Ciudad del Conocimiento, Carretera Pachuca-Tulancingo km 4.5, Colonia Carboneras. CP. 42184. Mineral de la Reforma, Hidalgo, México
| | - Mirandeli Bautista-Ávila
- Universidad Autónoma del Estado de Hidalgo. Área Académica de Farmacia, Instituto de Ciencias de la Salud. Ex-Hacienda la Concepción. San Agustín Tlaxiaca, Hidalgo, México
| | - Alma Elizabeth Cruz-Guerrero
- Universidad Autónoma Metropolitana, Unidad Iztapalapa. División de Ciencias Biológicas y de la Salud. Departamento de Biotecnología, Colonia Vicentina AP 09340, Ciudad de México, México
| | - Juan Francisco Flores-Aguilar
- Universidad Autónoma del Estado de Hidalgo, Área Académica de Química. Ciudad del Conocimiento, Carretera Pachuca-Tulancingo km 4.5, Colonia Carboneras. CP. 42184. Mineral de la Reforma, Hidalgo, México
| | - Luis Guillermo González-Olivares
- Universidad Autónoma del Estado de Hidalgo, Área Académica de Química. Ciudad del Conocimiento, Carretera Pachuca-Tulancingo km 4.5, Colonia Carboneras. CP. 42184. Mineral de la Reforma, Hidalgo, México
| |
Collapse
|
25
|
Cox AJ, Cripps AW, Taylor PA, Fitton JH, West NP. Fucoidan Supplementation Restores Fecal Lysozyme Concentrations in High-Performance Athletes: A Pilot Study. Mar Drugs 2020; 18:md18080412. [PMID: 32759709 PMCID: PMC7460205 DOI: 10.3390/md18080412] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 06/30/2020] [Accepted: 08/03/2020] [Indexed: 12/14/2022] Open
Abstract
Nutritional strategies to help promote immune competence are of particular interest for a range of population groups. This study aimed to assess the potential impacts of fucoidan, a seaweed-derived bioactive polysaccharide, on gut markers of immunity and inflammation. A group of professional team-sport athletes were selected for inclusion in the study given the recognized potential for intense physical activity to induce alterations in immune function. A retrospective analysis was performed on stored fecal samples which had been collected from professional team-sport athletes (n = 22) and healthy adults (n = 11) before and after seven days of supplementation with fucoidan (Fucus vesiculosus/Undaria pinnatifida extract, 1 g/d). Fecal concentrations of calprotectin, secretory immunoglobulin A (sIgA) and lysozyme were determined using enzyme-linked immunosorbent assays. The supplement was well tolerated by participants with no adverse events reported. At baseline, fecal lysozyme concentrations were ~73% higher in the healthy adults compared to the professional athletes (p = 0.001). For the professional athletes, a significant (~45%) increase in fecal lysozyme was observed following the supplementation period (p = 0.001). These data suggest that fucoidan supplementation may have the potential to promote the secretion of antimicrobial peptides in specific population groups and contribute to the regulation of mucosal immune health.
Collapse
Affiliation(s)
- Amanda J. Cox
- School of Medical Science, Griffith University, Southport, QLD 4215, Australia;
- Menzies Health Institute Queensland, Griffith University, Southport, QLD 4215, Australia;
- Correspondence: ; Tel.: +61-7-5678-0898
| | - Allan W. Cripps
- Menzies Health Institute Queensland, Griffith University, Southport, QLD 4215, Australia;
- School of Medicine, Griffith University, Southport, QLD 4215, Australia
| | | | | | - Nicholas P. West
- School of Medical Science, Griffith University, Southport, QLD 4215, Australia;
- Menzies Health Institute Queensland, Griffith University, Southport, QLD 4215, Australia;
| |
Collapse
|
26
|
Salminen S, Stahl B, Vinderola G, Szajewska H. Infant Formula Supplemented with Biotics: Current Knowledge and Future Perspectives. Nutrients 2020; 12:E1952. [PMID: 32629970 PMCID: PMC7400136 DOI: 10.3390/nu12071952] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/27/2020] [Accepted: 06/28/2020] [Indexed: 02/06/2023] Open
Abstract
Breastfeeding is natural and the optimal basis of infant nutrition and development, with many benefits for maternal health. Human milk is a dynamic fluid fulfilling an infant's specific nutritional requirements and guiding the growth, developmental, and physiological processes of the infant. Human milk is considered unique in composition, and it is influenced by several factors, such as maternal diet and health, body composition, and geographic region. Human milk stands as a model for infant formula providing nutritional solutions for infants not able to receive enough mother's milk. Infant formulas aim to mimic the composition and functionality of human milk by providing ingredients reflecting those of the latest human milk insights, such as oligosaccharides, bacteria, and bacterial metabolites. The objective of this narrative review is to discuss the most recent developments in infant formula with a special focus on human milk oligosaccharides and postbiotics.
Collapse
Affiliation(s)
- Seppo Salminen
- Functional Foods Forum, Faculty of Medicine, University of Turku, 20520 Turku, Finland;
| | - Bernd Stahl
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands;
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Gabriel Vinderola
- Instituto de Lactología Industrial (INLAIN, UNL-CONICET), Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santiago del Estero 2829, Santa Fe 3000, Argentina;
| | - Hania Szajewska
- Department of Paediatrics at the Medical University of Warsaw, 02091 Warsaw, Poland
| |
Collapse
|
27
|
Quinn EM, Joshi L, Hickey RM. Symposium review: Dairy-derived oligosaccharides-Their influence on host-microbe interactions in the gastrointestinal tract of infants. J Dairy Sci 2020; 103:3816-3827. [PMID: 32089300 DOI: 10.3168/jds.2019-17645] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/18/2019] [Indexed: 12/13/2022]
Abstract
Oligosaccharides are the third most abundant component in human milk. It is widely accepted that they play several important protective, physiological, and biological roles, including selective growth stimulation of beneficial gut microbiota, inhibition of pathogen adhesion, and immune modulation. However, until recently, very few commercial products on the market have capitalized on these functions. This is mainly because the quantities of human milk oligosaccharides required for clinical trials have been unavailable. Recently, clinical studies have tested the potential beneficial effects of feeding infants formula containing 2'-fucosyllactose, which is the most abundant oligosaccharide in human milk. These studies have opened this field for further well-designed studies, which are required to fully understand the role of human milk oligosaccharides. However, one of the most striking features of human milk is its diversity of oligosaccharides, with over 200 identified to date. It may be that a mixture of oligosaccharides is even more beneficial to infants than a single structure. For this reason, the milk of domestic animals has become a focal point in recent years as an alternative source of complex oligosaccharides with associated biological activity. This review will focus specifically on free oligosaccharides found in bovine and caprine milk and the biological roles associated with such structures. These dairy streams are ideal sources of oligosaccharides, given their wide availability and use in so many regularly consumed dairy products. The aim of this review was to provide an overview of research into the functional role of bovine and caprine milk oligosaccharides in host-microbial interactions in the gut and provide current knowledge related to the isolation of oligosaccharides as ingredients for incorporation in functional or medical foods.
Collapse
Affiliation(s)
- Erinn M Quinn
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland; Advanced Glycoscience Research Cluster, National Centre for Biomedical Engineering Science, National University of Ireland Galway, Galway H91 TK33, Ireland
| | - Lokesh Joshi
- Advanced Glycoscience Research Cluster, National Centre for Biomedical Engineering Science, National University of Ireland Galway, Galway H91 TK33, Ireland
| | - Rita M Hickey
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland.
| |
Collapse
|
28
|
Cerdó T, Diéguez E, Campoy C. Infant growth, neurodevelopment and gut microbiota during infancy: which nutrients are crucial? Curr Opin Clin Nutr Metab Care 2019; 22:434-441. [PMID: 31567222 DOI: 10.1097/mco.0000000000000606] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW To update the role of specific nutrients during infant development. RECENT FINDINGS Several bioactive nutrients such as long-chain polyunsaturated fatty acids (LC-PUFAs), iron, vitamins, proteins, or carbohydrates have been identified to exert an important role during the first 1000 days of life on infant growth, neurodevelopment, and gut microbiota establishment and maturation. LC-PUFAs are structural constituents of the central nervous system (CNS), being essential in retinal development or hippocampal plasticity. Recently, components of the milk fat globule membrane (MFG) are being added to infant formulas because of their key role in infant's development. A high intake of proteins induces a faster weight gain during infancy which correlates with later obesity. Digestible carbohydrates provide glucose, crucial for an adequate functioning of CNS; nondigestible carbohydrates [e.g. human milk oligosaccharides (HMOs)] are the main carbon source for gut bacteria. Iron-deficiency anemia during infancy has been associated with alterations of mental and psychomotor development. Folate metabolism, closely related to vitamins B6 and B12, controls epigenetic changes, whereas inadequate status of vitamin D affects bone development, but may also increase intestinal permeability and alter gut microbiota composition. SUMMARY LC-PUFAs, proteins, carbohydrates, iron, and vitamins during early life are critical for infant's growth, neurodevelopment, and the establishment and functioning of gut microbiota.
Collapse
Affiliation(s)
- Tomás Cerdó
- Department of Paediatrics, School of Medicine
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada
- BioHealth Research Institute (Ibs), Granada, Health Sciences Technological Park
- Neurosciences Institute, Biomedical Research Centre, University of Granada, Granada
| | - Estefanía Diéguez
- Department of Paediatrics, School of Medicine
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada
| | - Cristina Campoy
- Department of Paediatrics, School of Medicine
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada
- BioHealth Research Institute (Ibs), Granada, Health Sciences Technological Park
- Neurosciences Institute, Biomedical Research Centre, University of Granada, Granada
- Spanish Network of Biomedical Research in Epidemiology and Public Health (CIBERESP), Granada's node, Carlos III Health Institute of Health Carlos III, Madrid
- Brain, Behavior and Health Excellence Research Unit, (SC2). University of Granada, Granada, Spain
| |
Collapse
|
29
|
|
30
|
Pitt J, Chan M, Gibson C, Hasselwander O, Lim A, Mukerji P, Mukherjea R, Myhre A, Sarela P, Tenning P, Himmelstein MW, Roper JM. Safety assessment of the biotechnologically produced human-identical milk oligosaccharide 3-Fucosyllactose (3-FL). Food Chem Toxicol 2019; 134:110818. [PMID: 31533061 DOI: 10.1016/j.fct.2019.110818] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/09/2019] [Accepted: 09/12/2019] [Indexed: 10/26/2022]
Abstract
3-Fucosyllactose (3-FL), a highly abundant complex carbohydrate in human breast milk, functions as a prebiotic promoting early microbial colonization of the gut, increasing pathogen resistance and modulating immune responses. To investigate potential health benefits, 3-FL was produced by fermentation using a genetically modified E. coli K12 strain. The safety assessment of 3-FL included acute oral toxicity, in vitro and in vivo assessment of genetic toxicity, and a subchronic rodent feeding study. 3-FL was not acutely toxic at 5000 mg/kg bw, and there was no evidence of genetic toxicity in the bacterial reverse mutation test and chromosomal aberration assay. There was a repeatable statistically-significant trend in the 4-h S9-activated test conditions in the in vitro micronucleus assay; the confirmatory in vivo mouse micronucleus study was negative at all doses. Dietary subchronic exposure of rats to 3-FL (5% and 10%) did not produce any statistical or biologically-relevant differences in growth, food intake or efficiency, clinical observations, or clinical or anatomic pathology changes at average daily intakes of 5.98 and 7.27 g/kg bw/day for males and females, respectively. The weight of evidence from these studies support the safe use of 3-FL produced using biotechnology as a nutritional ingredient in foods.
Collapse
Affiliation(s)
- J Pitt
- DuPont Nutrition and Biosciences, 1801 Larkin Center Drive, Midland, MI, 48674, USA.
| | - M Chan
- Corteva™, Agriscience™, P.O. Box 30, Newark, DE, 19714, USA
| | - C Gibson
- Total Pathology Solutions, LLC, Kennett Square, PA, 19348, USA
| | - O Hasselwander
- DuPont Nutrition and Biosciences, 43 London Road, Reigate, Surrey, RH2 9PW, UK
| | - A Lim
- DuPont Nutrition and Biosciences, 200 Powder Mill Road, Wilmington, DE, 19803, USA
| | - P Mukerji
- Corteva™, Agriscience™, P.O. Box 30, Newark, DE, 19714, USA
| | - R Mukherjea
- DuPont Nutrition and Biosciences, 4300 Duncan Ave, St. Louis MO, 63110, USA
| | - A Myhre
- Corteva™, Agriscience™, P.O. Box 30, Newark, DE, 19714, USA
| | - P Sarela
- DuPont Nutrition and Biosciences, Sokeritehtaanti 20, FI- 02460, Kantvik, FI, USA
| | - P Tenning
- DuPont Nutrition and Biosciences, Langebrogade 1, DK-1411, Copenhagen K, DE, USA
| | | | - J M Roper
- Corteva™, Agriscience™, P.O. Box 30, Newark, DE, 19714, USA
| |
Collapse
|
31
|
Hegar B, Wibowo Y, Basrowi RW, Ranuh RG, Sudarmo SM, Munasir Z, Atthiyah AF, Widodo AD, Supriatmo, Kadim M, Suryawan A, Diana NR, Manoppo C, Vandenplas Y. The Role of Two Human Milk Oligosaccharides, 2'-Fucosyllactose and Lacto-N-Neotetraose, in Infant Nutrition. Pediatr Gastroenterol Hepatol Nutr 2019; 22:330-340. [PMID: 31338308 PMCID: PMC6629589 DOI: 10.5223/pghn.2019.22.4.330] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/01/2019] [Accepted: 02/12/2019] [Indexed: 02/07/2023] Open
Abstract
Human breast milk contains numerous biomolecules. Human milk oligosaccharides (HMOs) are the third most abundant component of breast milk, after lactose and lipids. Amongst the synthetized HMOs, 2'-fucosyllactose (2'-FL) and lacto-N-neotetraose (LNnT) are widely studied and are considered safe for infant nutrition. Several studies have reported the health benefits of HMOs, which include modulation of the intestinal microbiota, anti-adhesive effect against pathogens, modulation of the intestinal epithelial cell response, and development of the immune system. The amount and diversity of HMOs are determined by the genetic background of the mothers (HMO secretors or non-secretors). The non-secretor mothers secrete lower HMOs than secretor mothers. The breastfed infants of secretor mothers gain more health benefit than those of non-secretor mothers. In conclusion, supplementation of infant formula with 2'-FL and LNnT is a promising innovation for infant nutrition.
Collapse
Affiliation(s)
- Badriul Hegar
- Cipto Mangunkusumo Hospital, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | | | | | - Reza Gunadi Ranuh
- Soetomo Hospital, Faculty of Medicine University Airlangga, Surabaya, Indonesia
| | | | - Zakiudin Munasir
- Cipto Mangunkusumo Hospital, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | | | | | - Supriatmo
- Adam Malik Hospital, Sumatera Utara, Indonesia
| | - Muzal Kadim
- Cipto Mangunkusumo Hospital, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Ahmad Suryawan
- Soetomo Hospital, Faculty of Medicine University Airlangga, Surabaya, Indonesia
| | | | | | - Yvan Vandenplas
- KidZ Health Castle, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
32
|
Restraint stress induced gut dysmotility is diminished by a milk oligosaccharide (2'-fucosyllactose) in vitro. PLoS One 2019; 14:e0215151. [PMID: 31017915 PMCID: PMC6481803 DOI: 10.1371/journal.pone.0215151] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 03/27/2019] [Indexed: 12/21/2022] Open
Abstract
Background Stress causes severe dysmotility in the mammalian gut. Almost all research done to date has concentrated on prevention of stress-induced altered gut motility but not on treatment. We had previously shown that intraluminal 2′FL could acutely moderate propulsive motility in isolated mouse colonic segments. Because 2′FL appeared to modulate enteric nervous system dependent motility, we wondered if the oligosaccharide could reverse the effects of prior restraint stress, ex vivo. We tested whether 2′FL could benefit the dysmotility of isolated jejunal and colonic segments from animals subjected to prior acute restraint stress. Methods Jejunal and colonic segments were obtained from male Swiss Webster mice that were untreated or subjected to 1 hour of acute restraint stress. Segments were perfused with Krebs buffer and propagating contractile clusters (PCC) digitally video recorded. 2′FL or β-lactose were added to the perfusate at a concentration of 1 mg/ml. Spatiotemporal maps were constructed from paired before and after treatment recordings, each consisting of 20 min duration and PCC analyzed for frequency, velocity and amplitude. Key Results Stress decreased propulsive motility in murine small intestine while increasing it in the colon. 2′FL in jejunum of previously stressed mice produced a 50% increase in PCC velocity (p = 0.0001), a 43% increase in frequency (p = 0.0002) and an insignificant decrease in peak amplitude. For stressed colon, 2′FL reduced the frequency by 23% (p = 0.017) and peak amplitude by 26% (p = 0.011), and was without effect on velocity. β-lactose had negligible or small treatment effects. Conclusions & Inferences We show that the prebiotic 2′FL may have potential as a treatment for acute stress-induced gut dysmotility, ex vivo, and that, as is the case for certain beneficial microbes, the mechanism occurs in the gut, likely via action on the enteric nervous system.
Collapse
|
33
|
Figueroa-Lozano S, de Vos P. Relationship Between Oligosaccharides and Glycoconjugates Content in Human Milk and the Development of the Gut Barrier. Compr Rev Food Sci Food Saf 2018; 18:121-139. [DOI: 10.1111/1541-4337.12400] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Susana Figueroa-Lozano
- Immunoendocrinology, Div. of Medical Biology, Dept. of Pathology and Medical Biology; Univ. of Groningen and University Medical Center Groningen; Groningen The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Div. of Medical Biology, Dept. of Pathology and Medical Biology; Univ. of Groningen and University Medical Center Groningen; Groningen The Netherlands
| |
Collapse
|
34
|
Wejryd E, Martí M, Marchini G, Werme A, Jonsson B, Landberg E, Abrahamsson TR. Low Diversity of Human Milk Oligosaccharides is Associated with Necrotising Enterocolitis in Extremely Low Birth Weight Infants. Nutrients 2018; 10:nu10101556. [PMID: 30347801 PMCID: PMC6213888 DOI: 10.3390/nu10101556] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 10/12/2018] [Accepted: 10/12/2018] [Indexed: 12/27/2022] Open
Abstract
Difference in human milk oligosaccharides (HMO) composition in breast milk may be one explanation why some preterm infants develop necrotizing enterocolitis (NEC) despite being fed exclusively with breast milk. The aim of this study was to measure the concentration of 15 dominant HMOs in breast milk during the neonatal period and investigate how their levels correlated to NEC, sepsis, and growth in extremely low birth weight (ELBW; <1000 g) infants who were exclusively fed with breast milk. Milk was collected from 91 mothers to 106 infants at 14 and 28 days and at postmenstrual week 36. The HMOs were analysed with high-performance anion-exchange chromatography with pulsed amperometric detection. The HMOs diversity and the levels of Lacto-N-difucohexaose I were lower in samples from mothers to NEC cases, as compared to non-NEC cases at all sampling time points. Lacto-N-difucohexaose I is only produced by secretor and Lewis positive mothers. There were also significant but inconsistent associations between 3′-sialyllactose and 6′-sialyllactose and culture-proven sepsis and significant, but weak correlations between several HMOs and growth rate. Our results suggest that the variation in HMO composition in breast milk may be an important factor explaining why exclusively breast milk fed ELBW infants develop NEC.
Collapse
Affiliation(s)
- Erik Wejryd
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden.
| | - Magalí Martí
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden.
| | - Giovanna Marchini
- Department of Neonatology, Karolinska University Hospital, 17176 Stockholm, Sweden.
- Department of Women´s and Children´s Health, Karolinska Insitute, 17177 Stockholm, Sweden.
| | - Anna Werme
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden.
| | - Baldvin Jonsson
- Department of Neonatology, Karolinska University Hospital, 17176 Stockholm, Sweden.
- Department of Women´s and Children´s Health, Karolinska Insitute, 17177 Stockholm, Sweden.
| | - Eva Landberg
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden.
- Department of Clinical Chemistry, Linköping University, 58185 Linköping, Sweden.
| | - Thomas R Abrahamsson
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden.
- Department of Pediatrics, Linköping University, 58183 Linköping, Sweden.
| |
Collapse
|
35
|
Oliveros E, Vázquez E, Barranco A, Ramírez M, Gruart A, Delgado-García JM, Buck R, Rueda R, Martín MJ. Sialic Acid and Sialylated Oligosaccharide Supplementation during Lactation Improves Learning and Memory in Rats. Nutrients 2018; 10:E1519. [PMID: 30332832 PMCID: PMC6212975 DOI: 10.3390/nu10101519] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/08/2018] [Accepted: 10/11/2018] [Indexed: 12/21/2022] Open
Abstract
Sialic acids (Sia) are postulated to improve cognitive abilities. This study evaluated Sia effects on rat behavior when administered in a free form as N-acetylneuraminic acid (Neu5Ac) or conjugated as 6'-sialyllactose (6'-SL). Rat milk contains Sia, which peaks at Postnatal Day 9 and drops to a minimum by Day 15. To bypass this Sia peak, a cohort of foster mothers was used to raise the experimental pups. A group of pups received a daily oral supplementation of Neu5Ac to mimic the amount naturally present in rat milk, and another group received the same molar amount of Sia as 6'-SL. The control group received water. After weaning, rats were submitted to behavioral evaluation. One year later, behavior was re-evaluated, and in vivo long-term potentiation (LTP) was performed. Brain samples were collected and analyzed at both ages. Adult rats who received Sia performed significantly better in the behavioral assessment and showed an enhanced LTP compared to controls. Within Sia groups, 6'-SL rats showed better scores in some cognitive outcomes compared to Neu5Ac rats. At weaning, an effect on polysialylated-neural cell adhesion molecule (PSA-NCAM) levels in the frontal cortex was only observed in 6'-SL fed rats. Providing Sia during lactation, especially as 6'-SL, improves memory and LTP in adult rats.
Collapse
Affiliation(s)
- Elena Oliveros
- R&D Abbott Nutrition, 18004 Granada, Spain.
- Doctoral programme in Biomedicine, School of Health Sciences, University of Granada, 18071 Granada, Spain.
| | | | | | | | - Agnes Gruart
- Division of Neurosciences, Pablo de Olavide University, 41013 Seville, Spain.
| | | | | | | | | |
Collapse
|
36
|
Human Milk Oligosaccharides: 2'-Fucosyllactose (2'-FL) and Lacto-N-Neotetraose (LNnT) in Infant Formula. Nutrients 2018; 10:nu10091161. [PMID: 30149573 PMCID: PMC6164445 DOI: 10.3390/nu10091161] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 08/09/2018] [Accepted: 08/14/2018] [Indexed: 12/27/2022] Open
Abstract
The authors reviewed the published evidence on the presence of oligosaccharides in human milk (HMO) and their benefits in in vitro and in vivo studies. The still limited data of trials evaluating the effect of mainly 2′-fucosyllactose (2′-FL) on the addition of some of HMOs to infant formula were also reviewed. PubMed was searched from January 1990 to April 2018. The amount of HMOs in mother’s milk is a dynamic process as it changes over time. Many factors, such as duration of lactation, environmental, and genetic factors, influence the amount of HMOs. HMOs may support immune function development and provide protection against infectious diseases directly through the interaction of the gut epithelial cells or indirectly through the modulation of the gut microbiota, including the stimulation of the bifidobacteria. The limited clinical data suggest that the addition of HMOs to infant formula seems to be safe and well tolerated, inducing a normal growth and suggesting a trend towards health benefits. HMOs are one of the major differences between cow’s milk and human milk, and available evidence indicates that these components do have a health promoting benefit. The addition of one or two of these components to infant formula is safe, and brings infant formula closer to human milk. More prospective, randomized trials in infants are need to evaluate the clinical benefit of supplementing infant formula with HMOs.
Collapse
|
37
|
Safety evaluation of a mixture of the human-identical milk oligosaccharides 2'-fucosyllactose and difucosyllactose. Food Chem Toxicol 2018; 120:552-565. [PMID: 30076915 DOI: 10.1016/j.fct.2018.07.054] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/25/2018] [Accepted: 07/28/2018] [Indexed: 12/24/2022]
Abstract
Human milk oligosaccharides (HMOs) are endogenous indigestible carbohydrates representing the largest compositional difference between human breastmilk and infant formula (IF). Two major HMOs in human breastmilk are 2'-fucosyllactose (2'-FL) and difucosyllactose (DFL); commercial IF can be supplemented with manufactured structurally identical versions of HMOs [known as human-identical milk oligosaccharides (HiMOs)] to better replicate the composition of human milk. As 2'-FL and DFL are always found together in human milk, a mixture of these HiMOs (2'-FL/DFL) has been proposed for use in IF and as a food ingredient. Safety assessment of 2'-FL/DFL included conduct of in vitro genotoxicity tests and a subchronic oral toxicity study. In the subchronic study, 2'-FL/DFL (8:1 ratio) was administered to neonatal rats at doses up to 5000 mg/kg body weight (bw)/day, once daily for 90 days, followed by a 4-week recovery period. A concurrent reference control group received 5000 mg/kg bw/day of an oligosaccharide already used in IF (fructooligosaccharide), for direct comparison with the high-dose 2'-FL/DFL group. No evidence of genotoxicity was observed. In the absence of compound-related adverse effects in the 90-day study, 5000 mg/kg bw/day was established as the no-observed-adverse-effect-level. These results support the use of 2'-FL/DFL in IF and as a food ingredient.
Collapse
|
38
|
Zehra S, Khambati I, Vierhout M, Mian MF, Buck R, Forsythe P. Human Milk Oligosaccharides Attenuate Antigen-Antibody Complex Induced Chemokine Release from Human Intestinal Epithelial Cell Lines. J Food Sci 2018; 83:499-508. [PMID: 29377120 DOI: 10.1111/1750-3841.14039] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/24/2017] [Accepted: 12/11/2017] [Indexed: 12/17/2022]
Abstract
There has been increased interest in the use of dietary ingredients, including prebiotics such as human-milk oligosaccharides (HMOs), as therapeutic strategies for food allergy. Understanding the mechanisms underlying the beneficial effects of HMOs is important to realizing their therapeutic potential. Here we demonstrate that the HMO, 6'-sialyllactose (6'SL) inhibited chemokine (IL-8 and CCL20) release from T-84 and HT-29 cells stimulated with antigen-antibody complex, TNFα or PGE2 ; an effect that was PPARγ dependent and associated with decreased activity of the transcription factors AP-1 and NFκB. In contrast, 2'-fucosyllactose (2'FL) selectively inhibited CCL20 release in response to antigen antibody complex in a PPARγ independent manner. This study reinforces the concept that structurally different oligosaccharides have distinct biological activities and identifies, for the first time, that the HMOs, 6'SL, and 2'FL, modulate human epithelial cell responses related to allergic disease. These findings encourage further investigation of the therapeutic potential of specific HMOs in food allergy. PRACTICAL APPLICATION This study provides evidence for direct effects of HMOs in addition to their prebiotic role and demonstrates, for the first time, modulation of Ag-IgE complex activation of human epithelial cells that may have important implications for food-allergy. The study also reinforces the concept that structurally different oligosaccharides have distinct biological activities. In determining the composition of infant formula, addition of oligosaccharides with specific structures may provide direct modulation of immune responses and potentially attenuate symptoms or development of food allergy.
Collapse
Affiliation(s)
- Sehrish Zehra
- Dept. of Medicine, Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada
| | - Ibrahim Khambati
- Dept. of Medicine, Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada
| | - Megan Vierhout
- Dept. of Medicine, Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada
| | - M Firoz Mian
- Dept. of Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada
| | - Rachael Buck
- Abbott Nutrition: a Division of Abbott Laboratories, Columbus, Ohio, U.S.A
| | - Paul Forsythe
- Dept. of Medicine, Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada.,The Firestone Inst. for Respiratory Research, McMaster Univ., Hamilton, Ont., Canada
| |
Collapse
|
39
|
Turroni F, Milani C, Duranti S, Ferrario C, Lugli GA, Mancabelli L, van Sinderen D, Ventura M. Bifidobacteria and the infant gut: an example of co-evolution and natural selection. Cell Mol Life Sci 2018; 75:103-118. [PMID: 28983638 PMCID: PMC11105234 DOI: 10.1007/s00018-017-2672-0] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 09/29/2017] [Indexed: 12/16/2022]
Abstract
Throughout the human life, the gut microbiota interacts with us in a number of different ways, thereby influencing our health status. The acquisition of such an interactive gut microbiota commences at birth. Medical and environmental factors including diet, antibiotic exposure and mode of delivery are major factors that shape the composition of the microbial communities in the infant gut. Among the most abundant members of the infant microbiota are species belonging to the Bifidobacterium genus, which are believed to confer beneficial effects upon their host. Bifidobacteria may be acquired directly from the mother by vertical transmission and their persistence in the infant gut is associated with their saccharolytic activity toward glycans that are abundant in the infant gut. Here, we discuss the establishment of the infant gut microbiota and the contribution of bifidobacteria to this early life microbial consortium.
Collapse
Affiliation(s)
- Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Sabrina Duranti
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Chiara Ferrario
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Leonardo Mancabelli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy.
- Microbiome Research Hub, University of Parma, Parma, Italy.
| |
Collapse
|
40
|
Milani C, Duranti S, Bottacini F, Casey E, Turroni F, Mahony J, Belzer C, Delgado Palacio S, Arboleya Montes S, Mancabelli L, Lugli GA, Rodriguez JM, Bode L, de Vos W, Gueimonde M, Margolles A, van Sinderen D, Ventura M. The First Microbial Colonizers of the Human Gut: Composition, Activities, and Health Implications of the Infant Gut Microbiota. Microbiol Mol Biol Rev 2017; 81:e00036-17. [PMID: 29118049 PMCID: PMC5706746 DOI: 10.1128/mmbr.00036-17] [Citation(s) in RCA: 1054] [Impact Index Per Article: 131.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The human gut microbiota is engaged in multiple interactions affecting host health during the host's entire life span. Microbes colonize the neonatal gut immediately following birth. The establishment and interactive development of this early gut microbiota are believed to be (at least partially) driven and modulated by specific compounds present in human milk. It has been shown that certain genomes of infant gut commensals, in particular those of bifidobacterial species, are genetically adapted to utilize specific glycans of this human secretory fluid, thus representing a very intriguing example of host-microbe coevolution, where both partners are believed to benefit. In recent years, various metagenomic studies have tried to dissect the composition and functionality of the infant gut microbiome and to explore the distribution across the different ecological niches of the infant gut biogeography of the corresponding microbial consortia, including those corresponding to bacteria and viruses, in healthy and ill subjects. Such analyses have linked certain features of the microbiota/microbiome, such as reduced diversity or aberrant composition, to intestinal illnesses in infants or disease states that are manifested at later stages of life, including asthma, inflammatory bowel disease, and metabolic disorders. Thus, a growing number of studies have reported on how the early human gut microbiota composition/development may affect risk factors related to adult health conditions. This concept has fueled the development of strategies to shape the infant microbiota composition based on various functional food products. In this review, we describe the infant microbiota, the mechanisms that drive its establishment and composition, and how microbial consortia may be molded by natural or artificial interventions. Finally, we discuss the relevance of key microbial players of the infant gut microbiota, in particular bifidobacteria, with respect to their role in health and disease.
Collapse
Affiliation(s)
- Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Sabrina Duranti
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Francesca Bottacini
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Eoghan Casey
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Jennifer Mahony
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Susana Delgado Palacio
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Silvia Arboleya Montes
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Leonardo Mancabelli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Juan Miguel Rodriguez
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Lars Bode
- Department of Pediatrics and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California-San Diego, La Jolla, California, USA
| | - Willem de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- Department of Bacteriology & Immunology, RPU Immunobiology, University of Helsinki, Helsinki, Finland
| | - Miguel Gueimonde
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Abelardo Margolles
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| |
Collapse
|
41
|
Henrick BM, Yao XD, Nasser L, Roozrogousheh A, Rosenthal KL. Breastfeeding Behaviors and the Innate Immune System of Human Milk: Working Together to Protect Infants against Inflammation, HIV-1, and Other Infections. Front Immunol 2017; 8:1631. [PMID: 29238342 PMCID: PMC5712557 DOI: 10.3389/fimmu.2017.01631] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/09/2017] [Indexed: 12/21/2022] Open
Abstract
The majority of infants’ breastfeeding from their HIV-infected mothers do not acquire HIV-1 infection despite exposure to cell-free virus and cell-associated virus in HIV-infected breast milk. Paradoxically, exclusive breastfeeding regardless of the HIV status of the mother has led to a significant decrease in mother-to-child transmission (MTCT) compared with non-exclusive breastfeeding. Although it remains unclear how these HIV-exposed infants remain uninfected despite repeated and prolonged exposure to HIV-1, the low rate of transmission is suggestive of a multitude of protective, short-lived bioactive innate immune factors in breast milk. Indeed, recent studies of soluble factors in breast milk shed new light on mechanisms of neonatal HIV-1 protection. This review highlights the role and significance of innate immune factors in HIV-1 susceptibility and infection. Prevention of MTCT of HIV-1 is likely due to multiple factors, including innate immune factors such as lactoferrin and elafin among many others. In pursuing this field, our lab was the first to show that soluble toll-like receptor 2 (sTLR2) directly inhibits HIV infection, integration, and inflammation. More recently, we demonstrated that sTLR2 directly binds to selective HIV-1 proteins, including p17, gp41, and p24, leading to significantly reduced NFκB activation, interleukin-8 production, CCR5 expression, and HIV infection in a dose-dependent manner. Thus, a clearer understanding of soluble milk-derived innate factors with known antiviral functions may provide new therapeutic insights to reduce vertical HIV-1 transmission and will have important implications for protection against HIV-1 infection at other mucosal sites. Furthermore, innate bioactive factors identified in human milk may serve not only in protecting infants against infections and inflammation but also the elderly; thus, opening the door for novel innate immune therapeutics to protect newborns, infants, adults, and the elderly.
Collapse
Affiliation(s)
- Bethany M Henrick
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States.,Foods for Health Institute, University of California, Davis, Davis, CA, United States
| | - Xiao-Dan Yao
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Laila Nasser
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Ava Roozrogousheh
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Kenneth L Rosenthal
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
42
|
Kononova SV. How Fucose of Blood Group Glycotopes Programs Human Gut Microbiota. BIOCHEMISTRY. BIOKHIMIIA 2017; 82:973-989. [PMID: 28988527 DOI: 10.1134/s0006297917090012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Formation of appropriate gut microbiota is essential for human health. The first two years of life is the critical period for this process. Selection of mutualistic microorganisms of the intestinal microbiota is controlled by the FUT2 and FUT3 genes that encode fucosyltransferases, enzymes responsible for the synthesis of fucosylated glycan structures of mucins and milk oligosaccharides. In this review, the mechanisms of the selection and maintenance of intestinal microorganisms that involve fucosylated oligosaccharides of breast milk and mucins of the newborn's intestine are described. Possible reasons for the use of fucose, and not sialic acid, as the major biological signal for the selection are also discussed.
Collapse
Affiliation(s)
- S V Kononova
- Institute of Protein Research, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| |
Collapse
|
43
|
Thongaram T, Hoeflinger JL, Chow J, Miller MJ. Human milk oligosaccharide consumption by probiotic and human-associated bifidobacteria and lactobacilli. J Dairy Sci 2017; 100:7825-7833. [PMID: 28780103 DOI: 10.3168/jds.2017-12753] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 06/14/2017] [Indexed: 01/04/2023]
Abstract
Human milk contains high concentrations of nondigestible complex oligosaccharides (human milk oligosaccharides; HMO) that reach the colon and are subsequently fermented by the infant gut microbiota. Using a high-throughput, low-volume growth determination, we evaluated the ability of 12 lactobacilli and 12 bifidobacteria strains, including several commercial probiotics, to ferment HMO and their constituent monomers. Of the 24 strains tested, only Bifidobacterium longum ssp. infantis ATCC 15697 and Bifidobacterium infantis M-63 were able to ferment 3'-sialyllactose, 6'-sialyllactose, 2'-fucosyllactose, and 3'-fucosyllactose. Bifidobacterium infantis M-63 degraded almost 90% of the 2'-fucosyllactose but left most of the fucose in the supernatant, as detected by HPLC. Among bifidobacteria, only the B. infantis strains and Bifidobacterium breve ATCC 15700 were able to ferment lacto-N-neotetraose (LNnT). Among lactobacilli, Lactobacillus acidophilus NCFM was found to be the most efficient at utilizing LNnT. The extracellular β-galactosidase (lacL, LBA1467) of L. acidophilus NCFM cleaves the terminal galactose of LNnT for growth, leaving lacto-N-triose II in the media as detected by HPLC. Inactivation of lacL abolishes growth of L. acidophilus NCFM on LNnT. These results contribute to our knowledge of HMO-microbe interactions and demonstrate the potential for synbiotic combinations of pre- and probiotics.
Collapse
Affiliation(s)
- Taksawan Thongaram
- Department of Food Science and Human Nutrition, University of Illinois, Urbana 61801
| | - Jennifer L Hoeflinger
- Department of Food Science and Human Nutrition, University of Illinois, Urbana 61801
| | | | - Michael J Miller
- Department of Food Science and Human Nutrition, University of Illinois, Urbana 61801.
| |
Collapse
|
44
|
Major human milk oligosaccharides are absorbed into the systemic circulation after oral administration in rats. Br J Nutr 2017; 117:237-247. [DOI: 10.1017/s0007114516004554] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
AbstractHuman milk oligosaccharides (HMO) are involved in many biological functions influencing infant health. Although HMO act locally at the intestine, recent evidence has demonstrated that HMO are partially incorporated into the systemic circulation of breast-fed infants. In the last few years, a large amount of research has been conducted using preclinical models to uncover new biological functions of HMO. The aim of this study was to evaluate the absorption and urine excretion of HMO in rats. We administered a single oral dose of the following HMO: 2'-fucosyllactose (2'-FL), 6'-sialyllactose and lacto-N-neotetraose at different concentrations to adult rats. The time course of absorption of HMO into the bloodstream and their appearance in urine was studied. Our results showed that rats, similar to human infants, are able to effectively absorb a portion of HMO from the intestine into plasma and to excrete them in urine. On the basis of this, we also conducted a specific kinetic absorption study with 2'-FL, the most predominant HMO in human milk, in 9–11-d-old rat pups. Our results confirmed that a significant amount of 2'-FL was absorbed into the systemic circulation and subsequently excreted in urine during lactation in rats in a dose-depended manner. We also found basal levels of these HMO in plasma and urine of adult rats as well as rat pups as a natural result of nursing. Our data suggest that the rat may be a useful preclinical model that provides new insights into the metabolism and functions of HMO.
Collapse
|
45
|
Vazquez E, Barranco A, Ramirez M, Gruart A, Delgado-Garcia JM, Jimenez ML, Buck R, Rueda R. Dietary 2'-Fucosyllactose Enhances Operant Conditioning and Long-Term Potentiation via Gut-Brain Communication through the Vagus Nerve in Rodents. PLoS One 2016; 11:e0166070. [PMID: 27851789 PMCID: PMC5113009 DOI: 10.1371/journal.pone.0166070] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/22/2016] [Indexed: 01/06/2023] Open
Abstract
2´-fucosyllactose (2´-FL) is an abundant human milk oligosaccharide (HMO) in human milk with diverse biological effects. We recently reported ingested 2´-FL stimulates central nervous system (CNS) function, such as hippocampal long term potentiation (LTP) and learning and memory in rats. Conceivably the effect of 2´-FL on CNS function may be via the gut-brain axis (GBA), specifically the vagus nerve, and L-fucose (Fuc) may play a role. This study had two aims: (1) determine if the effect of ingested 2´-FL on the modulation of CNS function is dependent on the integrity of the molecule; and (2) confirm if oral 2´-FL modified hippocampal LTP and associative learning related skills in rats submitted to bilateral subdiaphragmatic vagotomy. Results showed that 2´-FL but not Fuc enhanced LTP, and vagotomy inhibited the effects of oral 2´-FL on LTP and associative learning related paradigms. Taken together, the data show that dietary 2´-FL but not its Fuc moiety affects cognitive domains and improves learning and memory in rats. This effect is dependent on vagus nerve integrity, suggesting GBA plays a role in 2´-FL-mediated cognitive benefits.
Collapse
Affiliation(s)
- Enrique Vazquez
- Strategic R&D Department, Abbott Nutrition, Granada, 18004, Spain
- * E-mail:
| | | | - Maria Ramirez
- Strategic R&D Department, Abbott Nutrition, Granada, 18004, Spain
| | - Agnes Gruart
- Division of Neurosciences, Pablo de Olavide University, Seville, 41013, Spain
| | | | - Maria L. Jimenez
- Strategic R&D Department, Abbott Nutrition, Granada, 18004, Spain
| | - Rachael Buck
- Strategic R&D Department, Abbott Nutrition, Columbus, OH, United States of America
| | - Ricardo Rueda
- Strategic R&D Department, Abbott Nutrition, Granada, 18004, Spain
| |
Collapse
|
46
|
Bienenstock J, Kunze W, Forsythe P. The Microbiome–Gut–Brain Axis and the Consequences of Infection and Dysbiosis. ACTA ACUST UNITED AC 2016. [DOI: 10.1038/ajgsup.2016.12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
47
|
Noll AJ, Gourdine JP, Yu Y, Lasanajak Y, Smith DF, Cummings RD. Galectins are human milk glycan receptors. Glycobiology 2016; 26:655-69. [PMID: 26747425 PMCID: PMC4847615 DOI: 10.1093/glycob/cww002] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 12/21/2015] [Accepted: 01/05/2016] [Indexed: 12/12/2022] Open
Abstract
The biological recognition of human milk glycans (HMGs) is poorly understood. Because HMGs are rich in galactose we explored whether they might interact with human galectins, which bind galactose-containing glycans and are highly expressed in epithelial cells and other cell types. We screened a number of human galectins for their binding to HMGs on a shotgun glycan microarray consisting of 247 HMGs derived from human milk, as well as to a defined HMG microarray. Recombinant human galectins (hGal)-1, -3, -4, -7, -8 and -9 bound selectively to glycans, with each galectin recognizing a relatively unique binding motif; by contrast hGal-2 did not recognize HMGs, but did bind to the human blood group A Type 2 determinants on other microarrays. Unlike other galectins, hGal-7 preferentially bound to glycans expressing a terminal Type 1 (Galβ1-3GlcNAc) sequence, a motif that had eluded detection on non-HMG glycan microarrays. Interactions with HMGs were confirmed in a solution setting by isothermal titration microcalorimetry and hapten inhibition experiments. These results demonstrate that galectins selectively bind to HMGs and suggest the possibility that galectin-HMG interactions may play a role in infant immunity.
Collapse
Affiliation(s)
- Alexander J Noll
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA, USA
| | | | - Ying Yu
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA, USA
| | - Yi Lasanajak
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA, USA
| | - David F Smith
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA, USA
| | - Richard D Cummings
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA, USA Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11087-3 Blackfan Circle, Boston, MA 02115, USA
| |
Collapse
|
48
|
Noll AJ, Yu Y, Lasanajak Y, Duska-McEwen G, Buck RH, Smith DF, Cummings RD. Human DC-SIGN binds specific human milk glycans. Biochem J 2016; 473:1343-53. [PMID: 26976925 PMCID: PMC4875834 DOI: 10.1042/bcj20160046] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/14/2016] [Indexed: 01/12/2023]
Abstract
Human milk glycans (HMGs) are prebiotics, pathogen receptor decoys and regulators of host physiology and immune responses. Mechanistically, human lectins (glycan-binding proteins, hGBP) expressed by dendritic cells (DCs) are of major interest, as these cells directly contact HMGs. To explore such interactions, we screened many C-type lectins and sialic acid-binding immunoglobulin-like lectins (Siglecs) expressed by DCs for glycan binding on microarrays presenting over 200 HMGs. Unexpectedly, DC-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) showed robust binding to many HMGs, whereas other C-type lectins failed to bind, and Siglec-5 and Siglec-9 showed weak binding to a few glycans. By contrast, most hGBP bound to multiple glycans on other microarrays lacking HMGs. An α-linked fucose residue was characteristic of HMGs bound by DC-SIGN. Binding of DC-SIGN to the simple HMGs 2'-fucosyl-lactose (2'-FL) and 3-fucosyl-lactose (3-FL) was confirmed by flow cytometry to beads conjugated with 2'-FL or 3-FL, as well as the ability of the free glycans to inhibit DC-SIGN binding. 2'-FL had an IC50 of ∼1 mM for DC-SIGN, which is within the physiological concentration of 2'-FL in human milk. These results demonstrate that DC-SIGN among the many hGBP expressed by DCs binds to α-fucosylated HMGs, and suggest that such interactions may be important in influencing immune responses in the developing infant.
Collapse
Affiliation(s)
- Alexander J Noll
- The Glycomics Center, Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, U.S.A. Program in Microbiology and Molecular Genetics, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA 30322, U.S.A
| | - Ying Yu
- The Glycomics Center, Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, U.S.A
| | - Yi Lasanajak
- The Glycomics Center, Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, U.S.A
| | | | - Rachael H Buck
- Abbott Nutrition, Global Discovery R&D, Columbus, OH 43215, U.S.A
| | - David F Smith
- The Glycomics Center, Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, U.S.A
| | - Richard D Cummings
- The Glycomics Center, Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, U.S.A.
| |
Collapse
|
49
|
Forsythe P, Kunze W, Bienenstock J. Moody microbes or fecal phrenology: what do we know about the microbiota-gut-brain axis? BMC Med 2016; 14:58. [PMID: 27090095 PMCID: PMC4836158 DOI: 10.1186/s12916-016-0604-8] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 03/18/2016] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION The microbiota-gut-brain axis is a term that is commonly used and covers a broad set of functions and interactions between the gut microbiome, endocrine, immune and nervous systems and the brain. The field is not much more than a decade old and so large holes exist in our knowledge. DISCUSSION At first sight it appears gut microbes are largely responsible for the development, maturation and adult function of the enteric nervous system as well as the blood brain barrier, microglia and many aspects of the central nervous system structure and function. Given the state of the art in this exploding field and the hopes, as well as the skepticism, which have been engendered by its popular appeal, we explore recent examples of evidence in rodents and data derived from studies in humans, which offer insights as to pathways involved. Communication between gut and brain depends on both humoral and nervous connections. Since these are bi-directional and occur through complex communication pathways, it is perhaps not surprising that while striking observations have been reported, they have often either not yet been reproduced or their replication by others has not been successful. CONCLUSIONS We offer critical and cautionary commentary on the available evidence, and identify gaps in our knowledge that need to be filled so as to achieve translation, where possible, into beneficial application in the clinical setting.
Collapse
Affiliation(s)
- Paul Forsythe
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada. .,McMaster Brain-Body Institute, St. Joseph's Healthcare, Hamilton, Ontario, Canada. .,Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, Ontario, Canada.
| | - Wolfgang Kunze
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada.,McMaster Brain-Body Institute, St. Joseph's Healthcare, Hamilton, Ontario, Canada
| | - John Bienenstock
- Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.,McMaster Brain-Body Institute, St. Joseph's Healthcare, Hamilton, Ontario, Canada
| |
Collapse
|
50
|
Abstract
The microbiota has recently been recognized as a driver of health that affects the immune, nervous, and metabolic systems. This influence is partially exerted through the metabolites produced, which may be relevant for optimal infant development and health. The gut microbiota begins developing early in life, and this initial colonization is remarkably important because it may influence long-term microbiota composition and activity. Considering that the microbiome may play a key role in health and disease, maintaining a protective microbiota could be critical in preventing dysbiosis-related diseases such as allergies, autoimmunity disorders, and metabolic syndrome. Breast milk and milk glycans in particular are thought to play a major role in shaping the early-life microbiota and promoting its development, thus affecting health. This review describes some of the effects the microbiota has on the host and discusses the role microbial metabolites play in shaping newborn health and development. We describe the gut microbiota structure and function during early life and the factors that determine its composition and hypothesize about the effects of human milk oligosaccharides and other prebiotic fibers on the neonatal microbiota.
Collapse
Affiliation(s)
| | - Maria J Martin
- Discovery R&D Department, Abbott Nutrition, Granada, Spain
| | | |
Collapse
|