101
|
Zhou J, Wang Y, Fan Q, Liu Y, Liu H, Yan J, Li M, Dong W, Li W. High levels of fucosylation and sialylation of milk N-glycans from mothers with gestational diabetes mellitus alter the offspring gut microbiome and immune balance in mice. FASEB J 2020; 34:3715-3731. [PMID: 31944389 DOI: 10.1096/fj.201901674r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 12/09/2019] [Accepted: 12/17/2019] [Indexed: 12/17/2022]
Abstract
Gestational diabetes mellitus (GDM) is significantly associated with allergen sensitization in early childhood, and this may influence the gut microbiome and immune system of the children. In addition to mother-to-child transmission of microbes, milk glycans play a pivotal role in shaping the gut microbiome of infants. A previous study has demonstrated alterations in the major milk N-glycans of mothers with GDM. However, the impact of these changes on the gut microbiome and immune response of the neonates has yet to be studied. Here, we aimed to compare the glycosylation levels of various milk glycans between normal and GDM mice, and to characterize the intestinal microbiome and immune responses of the offspring after weaning. We found that GDM mouse milk contained significantly higher concentrations of fucosylated and sialylated N-glycans than control mice, but there was no difference in the concentration of milk oligosaccharides between the groups. The differences in milk N-glycans had direct effects on the intestinal microbiome of the offspring, which in turn affected their immune response upon challenge with ovalbumin (OVA), with disruptions in the Th1/Th2 and Th17/Treg cell balances. This study lays the foundation for further research and development of specific nutritional care for the offspring of GDM mothers.
Collapse
Affiliation(s)
- Jiaorui Zhou
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Yue Wang
- Department of Biochemistry, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Qingjie Fan
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Yinhui Liu
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - He Liu
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Jingyu Yan
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Ming Li
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Weijie Dong
- Department of Biochemistry, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Wenzhe Li
- Department of Biochemistry, College of Basic Medical Science, Dalian Medical University, Dalian, China
| |
Collapse
|
102
|
Metabolomics reveals impact of seven functional foods on metabolic pathways in a gut microbiota model. J Adv Res 2020; 23:47-59. [PMID: 32071791 PMCID: PMC7016031 DOI: 10.1016/j.jare.2020.01.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/01/2020] [Accepted: 01/01/2020] [Indexed: 12/12/2022] Open
Abstract
Metabolomics was employed to assess 7 functional foods impact on gut microbiota. Insights regarding how functional foods alter gut metabolic pathways is presented. Increased GABA production was observed in polyphenol rich functional food. Purine alkaloids served as direct substrate in microbiota metabolism.
Functional food defined as dietary supplements that in addition to their nutritional values, can beneficially modulate body functions becomes more and more popular but the reaction of the intestinal microbiota to it is largely unknown. In order to analyse the impact of functional food on the microbiota itself it is necessary to focus on the physiology of the microbiota, which can be assessed in a whole by untargeted metabolomics. Obtaining a detailed description of the gut microbiota reaction to food ingredients can be a key to understand how these organisms regulate and bioprocess many of these food components. Extracts prepared from seven chief functional foods, namely green tea, black tea, Opuntia ficus-indica (prickly pear, cactus pear), black coffee, green coffee, pomegranate, and sumac were administered to a gut consortium culture encompassing 8 microbes which are resembling, to a large extent, the metabolic activities found in the human gut. Samples were harvested at 0.5 and 24 h post addition of functional food extract and from blank culture in parallel and analysed for its metabolites composition using gas chromatography coupled to mass spectrometry detection (GC-MS). A total of 131 metabolites were identified belonging to organic acids, alcohols, amino acids, fatty acids, inorganic compounds, nitrogenous compounds, nucleic acids, phenolics, steroids and sugars, with amino acids as the most abundant class in cultures. Considering the complexity of such datasets, multivariate data analyses were employed to classify samples and investigate how functional foods influence gut microbiota metabolisms. Results from this study provided a first insights regarding how functional foods alter gut metabolism through either induction or inhibition of certain metabolic pathways, i.e. GABA production in the presence of higher acidity induced by functional food metabolites such as polyphenols. Likewise, functional food metabolites i.e., purine alkaloids acted themselves as direct substrate in microbiota metabolism.
Collapse
Key Words
- BC, Black Coffee
- BT, Black Tea
- Chemometrics
- FI, Opuntia ficus-indica (prickly pear)
- Functional foods
- GC, Green Coffee
- GCMS
- GI, gastrointestinal
- GIT, gastrointestinal tract
- GT, Green Tea
- Gut microbiota
- Metabolomics
- POM, pomegranate (Punica granatum)
- SCFAs, short chain fatty acids
- SUM, sumac (Rhus coriaria)
Collapse
|
103
|
Sousa YR, Medeiros LB, Pintado MME, Queiroga RC. Goat milk oligosaccharides: Composition, analytical methods and bioactive and nutritional properties. Trends Food Sci Technol 2019. [DOI: 10.1016/j.tifs.2019.07.052] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
104
|
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
|
105
|
Salli K, Anglenius H, Hirvonen J, Hibberd AA, Ahonen I, Saarinen MT, Tiihonen K, Maukonen J, Ouwehand AC. The effect of 2'-fucosyllactose on simulated infant gut microbiome and metabolites; a pilot study in comparison to GOS and lactose. Sci Rep 2019; 9:13232. [PMID: 31520068 PMCID: PMC6744565 DOI: 10.1038/s41598-019-49497-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 08/27/2019] [Indexed: 01/01/2023] Open
Abstract
Human milk oligosaccharides (HMOs) shape gut microbiota during infancy by acting as fermentable energy source. Using a semi-continuous colon simulator, effect of an HMO, 2'-fucosyllactose (2'-FL), on composition of the infant microbiota and microbial metabolites was evaluated in comparison to galacto-oligosaccharide (GOS) and lactose and control without additional carbon source. Data was analysed according to faecal sample donor feeding type: breast-fed (BF) or formula-fed (FF), and to rate of 2'-FL fermentation: fast or slow. Variation was found between the simulations in the ability to utilise 2'-FL. The predominant phyla regulated by 2'-FL, GOS and lactose were significant increase in Firmicutes, numerical in Actinobacteria, and numerical decrease in Proteobacteria compared to control. Verrucomicrobia increased in FF accounted for Akkermansia, whereas in fast-fermenting simulations Actinobacteria increased with trend for higher Bifidobacterium, and Proteobacteria decrease accounted for Enterobacteriaceae. Short-chain fatty acids and lactic acid with 2'-FL were produced in intermediate levels being between ones generated by the control and GOS or lactose. In 2'-FL fast-fermenting group, acetic acid specifically increased with 2'-FL, whereas lactose and GOS also increased lactic acid. The results highlight specificity of 2'-FL as energy source for only certain microbes over GOS and lactose in the simulated gut model.
Collapse
Affiliation(s)
- Krista Salli
- DuPont Nutrition & Biosciences, Global Health & Nutrition Science, Kantvik, Finland.
| | - Heli Anglenius
- DuPont Nutrition & Biosciences, Global Health & Nutrition Science, Kantvik, Finland
| | - Johanna Hirvonen
- DuPont Nutrition & Biosciences, Global Health & Nutrition Science, Kantvik, Finland
| | - Ashley A Hibberd
- DuPont Nutrition & Biosciences, Genomics & Microbiome Science, Madison, WI, USA
| | | | - Markku T Saarinen
- DuPont Nutrition & Biosciences, Global Health & Nutrition Science, Kantvik, Finland
| | - Kirsti Tiihonen
- DuPont Nutrition & Biosciences, Global Health & Nutrition Science, Kantvik, Finland
| | - Johanna Maukonen
- DuPont Nutrition & Biosciences, Global Health & Nutrition Science, Kantvik, Finland
| | - Arthur C Ouwehand
- DuPont Nutrition & Biosciences, Global Health & Nutrition Science, Kantvik, Finland
| |
Collapse
|
106
|
Choi YH, Park BS, Seo JH, Kim BG. Biosynthesis of the human milk oligosaccharide 3-fucosyllactose in metabolically engineered Escherichia coli via the salvage pathway through increasing GTP synthesis and β-galactosidase modification. Biotechnol Bioeng 2019; 116:3324-3332. [PMID: 31478191 DOI: 10.1002/bit.27160] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/27/2019] [Accepted: 08/25/2019] [Indexed: 12/13/2022]
Abstract
3-Fucosyllactose (3-FL) is one of the major fucosylated oligosaccharides in human milk. Along with 2'-fucosyllactose (2'-FL), it is known for its prebiotic, immunomodulator, neonatal brain development, and antimicrobial function. Whereas the biological production of 2'-FL has been widely studied and made significant progress over the years, the biological production of 3-FL has been hampered by the low activity and insoluble expression of α-1,3-fucosyltransferase (FutA), relatively low abundance in human milk oligosaccharides compared with 2'-FL, and lower digestibility of 3-FL than 2'-FL by bifidobacteria. In this study, we report the gram-scale production of 3-FL using E. coli BL21(DE3). We previously generated the FutA quadruple mutant (mFutA) with four site mutations at S46F, A128N, H129E, Y132I, and its specific activity was increased by nearly 15 times compared with that of wild-type FutA owing to the increase in kcat and the decrease in Km . We overexpressed mFutA in its maximum expression level, which was achieved by the optimization of yeast extract concentration in culture media. We also overexpressed L-fucokinase/GDP- L-fucose pyrophosphorylase to increase the supply of GDP-fucose in the cytoplasm. To increase the mass of recombinant whole-cell catalysts, the host E. coli BW25113 was switched to E. coli BL21(DE3) because of the lower acetate accumulation of E. coli BL21(DE3) than that of E. coli BW25113. Finally, the lactose operon was modified by partially deleting the sequence of LacZ (lacZΔm15) for better utilization of D-lactose. Production using the lacZΔm15 mutant yielded 3-FL concentration of 4.6 g/L with the productivity of 0.076 g·L-1 ·hr-1 and the specific 3-FL yield of 0.5 g/g dry cell weight.
Collapse
Affiliation(s)
- Yun Hee Choi
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Republic of Korea.,Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Bum Seok Park
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea.,School of Chemical and Biological Engineering, Institute of Engineering Research, Seoul National University, Seoul, Republic of Korea
| | - Joo-Hyun Seo
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul, Republic of Korea
| | - Byung-Gee Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Republic of Korea.,Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea.,School of Chemical and Biological Engineering, Institute of Engineering Research, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
107
|
Kong C, Elderman M, Cheng L, de Haan BJ, Nauta A, de Vos P. Modulation of Intestinal Epithelial Glycocalyx Development by Human Milk Oligosaccharides and Non-Digestible Carbohydrates. Mol Nutr Food Res 2019; 63:e1900303. [PMID: 31140746 PMCID: PMC6771538 DOI: 10.1002/mnfr.201900303] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 03/26/2019] [Indexed: 12/11/2022]
Abstract
SCOPE The epithelial glycocalyx development is of great importance for microbial colonization. Human milk oligosaccharides (hMOs) and non-digestible carbohydrates (NDCs) may modulate glycocalyx development. METHODS AND RESULTS The effects of hMOs and NDCs on human gut epithelial cells (Caco2) are investigated by quantifying thickness and area coverage of adsorbed albumin, heparan sulfate (HS), and hyaluronic acid (HA) in the glycocalyx. Effects of hMOs (2'-FL and 3-FL) and NDCs [inulins with degrees of polymerization (DP) (DP3-DP10, DP10-DP60, DP30-DP60) and pectins with degrees of methylation (DM) (DM7, DM55, DM69)] are tested using immunofluorescence staining at 1 and 5 days stimulation. HMOs show a significant enhancing effect on glycocalyx development but effects are structure-dependent. 3-FL induces a stronger albumin adsorption and increases HS and HA stronger than 2'-FL. The DP3-DP10, DP30-60 inulins also increase glycocalyx development in a structure-dependent manner as DP3-DP10 selectively increases HS, while DP30-DP60 specifically increases HA. Pectins have less effects, and only increase albumin adsorption. CONCLUSION Here, it is shown that 2'-FL and 3-FL and inulins stimulate glycocalyx development in a structure-dependent fashion. This may contribute to formulation of effective hMO and NDC formulations in infant formulas to support microbial colonization and gut barrier function.
Collapse
Affiliation(s)
- Chunli Kong
- Immunoendocrinology GroupDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center Groningen, University of GroningenHanzeplein 19700 RBGroningenThe Netherlands
| | - Marlies Elderman
- Immunoendocrinology GroupDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center Groningen, University of GroningenHanzeplein 19700 RBGroningenThe Netherlands
| | - Lianghui Cheng
- Immunoendocrinology GroupDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center Groningen, University of GroningenHanzeplein 19700 RBGroningenThe Netherlands
| | - Bart J. de Haan
- Immunoendocrinology GroupDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center Groningen, University of GroningenHanzeplein 19700 RBGroningenThe Netherlands
| | - Arjen Nauta
- FrieslandCampinaStationsplein 43818 LEAmersfoortThe Netherlands
| | - Paul de Vos
- Immunoendocrinology GroupDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center Groningen, University of GroningenHanzeplein 19700 RBGroningenThe Netherlands
| |
Collapse
|
108
|
Faijes M, Castejón-Vilatersana M, Val-Cid C, Planas A. Enzymatic and cell factory approaches to the production of human milk oligosaccharides. Biotechnol Adv 2019; 37:667-697. [DOI: 10.1016/j.biotechadv.2019.03.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/22/2019] [Accepted: 03/23/2019] [Indexed: 12/15/2022]
|
109
|
Dey P. Gut microbiota in phytopharmacology: A comprehensive overview of concepts, reciprocal interactions, biotransformations and mode of actions. Pharmacol Res 2019; 147:104367. [PMID: 31344423 DOI: 10.1016/j.phrs.2019.104367] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/11/2019] [Accepted: 07/19/2019] [Indexed: 02/07/2023]
Abstract
The dynamic and delicate interactions amongst intestinal microbiota, metabolome and metabolism dictates human health and disease. In recent years, our understanding of gut microbial regulation of intestinal immunometabolic and redox homeostasis have evolved mainly out of in vivo studies associated with high-fat feeding induced metabolic diseases. Techniques utilizing fecal transplantation and germ-free mice have been instrumental in reproducibly demonstrating how the gut microbiota affects disease pathogenesis. However, the pillars of modern drug discovery i.e. evidence-based pharmacological studies critically lack focus on intestinal microflora. This is primarily due to targeted in vitro molecular-approaches at cellular-level that largely overlook the etiology of disease pathogenesis from the physiological perspective. Thus, this review aims to provide a comprehensive understanding of the key notions of intestinal microbiota and dysbiosis, and highlight the microbiota-phytochemical bidirectional interactions that affects bioavailability and bioactivity of parent phytochemicals and their metabolites. Potentially by focusing on the three major aspects of gut microbiota i.e. microbial abundance, diversity, and functions, I will discuss phytochemical-microbiota reciprocal interactions, biotransformation of phytochemicals and plant-derived drugs, and pre-clinical and clinical efficacies of herbal medicine on dysbiosis. Additionally, in relation to phytochemical pharmacology, I will briefly discuss the role of dietary-patterns associated with changes in microbial profiles and review pharmacological study models considering possible microbial effects. This review therefore, emphasize on the timely and critically needed evidence-based phytochemical studies focusing on gut microbiota and will provide newer insights for future pre-clinical and clinical phytopharmacological interventions.
Collapse
Affiliation(s)
- Priyankar Dey
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
110
|
Gao X, Lu Y, Wei M, Yang M, Zheng C, Wang C, Zhang Y, Huang L, Wang Z. Matrix-Assisted Laser Desorption/Ionization Time-of-Flight Mass Spectrometry Analysis of Human Milk Neutral and Sialylated Free Oligosaccharides Using Girard's Reagent P On-Target Derivatization. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:8958-8966. [PMID: 31334644 DOI: 10.1021/acs.jafc.9b02635] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The functional role of human milk oligosaccharides (HMOs) is closely associated with their type, composition, and structure. However, a detailed analysis of HMOs is difficult because neutral oligosaccharides (NHMOs) are mixed with sialylated oligosaccharides (SHMOs) in milk. Here, NHMOs were separated from SHMOs by DEAE-52 anion chromatography, and lactose was removed by graphite carbon solid-phase extraction. Lactose-free NHMOs were analyzed by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) based on Girard's reagent P on-target derivatization (GPOD), and SHMOs were analyzed by MALDI-TOF-MS following selective sialic acid derivatization and GPOD. Sixty-four oligosaccharides were detected: 36 NHMOs, of which 28 were fucosylated, and 28 SHMOs, of which 8 with α-2,3-linked monosialic acid, 2 with α-2,3-linked disialic acid, 10 with α-2,6-linked monosialic acid, 2 with α-2,6-linked disialic acid, and 5 with both α-2,3- and α-2,6-linked disialic acid. These findings provide the groundwork for further characterization of the structure and activity of HMOs.
Collapse
Affiliation(s)
| | | | | | | | - CaiXia Zheng
- The Second Affiliated Hospital of Xi'an Jiaotong University , Xi'an 710069 , China
| | | | | | | | | |
Collapse
|
111
|
Complete Genome Sequence of Kosakonia sp. Strain CCTCC M2018092, a Fucose-Rich Exopolysaccharide Producer. Microbiol Resour Announc 2019; 8:8/30/e00567-19. [PMID: 31346019 PMCID: PMC6658689 DOI: 10.1128/mra.00567-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Kosakonia sp. strain CCTCC M2018092 is a fucose-rich exopolysaccharide producer that was isolated from spring water in Chongqing, Southwest China. In this study, the whole-genome sequence and genetic characteristics of this strain were elucidated. This genome contained 4,789,478 bp, with a G+C content of 56.08% (excluding the plasmid). The genome information in this study will facilitate understanding of the mechanism of high yield of fucose-rich exopolysaccharide produced by Kosakonia sp. Kosakonia sp. strain CCTCC M2018092 is a fucose-rich exopolysaccharide producer that was isolated from spring water in Chongqing, Southwest China. In this study, the whole-genome sequence and genetic characteristics of this strain were elucidated. This genome contained 4,789,478 bp, with a G+C content of 56.08% (excluding the plasmid). The genome information in this study will facilitate understanding of the mechanism of high yield of fucose-rich exopolysaccharide produced by Kosakonia sp. CCTCC M2018092.
Collapse
|
112
|
Kuntz S, Kunz C, Borsch C, Vazquez E, Buck R, Reutzel M, Eckert GP, Rudloff S. Metabolic Fate and Distribution of 2´-Fucosyllactose: Direct Influence on Gut Microbial Activity but not on Brain. Mol Nutr Food Res 2019; 63:e1900035. [PMID: 31125176 PMCID: PMC6618057 DOI: 10.1002/mnfr.201900035] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/07/2019] [Indexed: 01/24/2023]
Abstract
SCOPE 2´-Fucosyllactose (2´FL) is an abundant oligosaccharide in human milk. It is hypothesized that its brain enrichment is associated with improved learning. Accumulation of 2´FL in organs, biological fluids, and feces is assessed in wild-type and germ-free mice. METHODS AND RESULTS 13 C-labelled 2´FL is applied to NMRI wild-type mice intravenously (0.2 g kg-1 ) or orally (1 g kg-1 ), while controls receive saline. Biological samples are collected (0.5-15 h) and 13 C-enrichment is measured by elemental analysis isotope ratio mass spectrometry (EA-IRMS). After oral application, 2´FL is primarily eliminated in the feces. 13 C-enrichment in organs including the brain follows the same pattern as in plasma with a maximum peak after 5 h. However, 13 C-enrichment is only detected when the 13 C-2´FL bolus reaches the colon. In contrast, in germ-free mice, the 13 C-bolus remains in the intestinal content and is expelled via the feces. Furthermore, intravenously applied 13 C-2´FL is eliminated via urine; no 13 C-enrichment of organs is observed, suggesting that intact 2´FL is not retained. CONCLUSIONS 13 C-enrichment in brain and other organs after oral application of 13 C-2´FL in wild-type mice indicates cleaved fucose or other gut microbial 2´FL metabolites may be incorporated, as opposed to intact 2´FL.
Collapse
Affiliation(s)
- Sabine Kuntz
- Institute of Nutritional SciencesJustus‐Liebig University Giessen35392GiessenGermany
| | - Clemens Kunz
- Institute of Nutritional SciencesJustus‐Liebig University Giessen35392GiessenGermany
| | - Christian Borsch
- Institute of Nutritional SciencesJustus‐Liebig University Giessen35392GiessenGermany
| | | | - Rachael Buck
- Discovery R&D, Abbott NutritionColumbusOH43219USA
| | - Martina Reutzel
- Institute of PharmacologyGoethe‐University Frankfurt60438Frankfurt am MainGermany
| | - Gunter Peter Eckert
- Institute of Nutritional SciencesJustus‐Liebig University Giessen35392GiessenGermany
- Institute of PharmacologyGoethe‐University Frankfurt60438Frankfurt am MainGermany
| | - Silvia Rudloff
- Institute of Nutritional SciencesJustus‐Liebig University Giessen35392GiessenGermany
- Department of PediatricsJustus‐Liebig University Giessen35392GiessenGermany
| |
Collapse
|
113
|
Novel Genes and Metabolite Trends in Bifidobacterium longum subsp. infantis Bi-26 Metabolism of Human Milk Oligosaccharide 2'-fucosyllactose. Sci Rep 2019; 9:7983. [PMID: 31138818 PMCID: PMC6538704 DOI: 10.1038/s41598-019-43780-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 05/01/2019] [Indexed: 12/11/2022] Open
Abstract
Human milk oligosaccharides (HMOs) function as prebiotics for beneficial bacteria in the developing gut, often dominated by Bifidobacterium spp. To understand the relationship between bifidobacteria utilizing HMOs and how the metabolites that are produced could affect the host, we analyzed the metabolism of HMO 2'-fucosyllactose (2'-FL) in Bifidobacterium longum subsp. infantis Bi-26. RNA-seq and metabolite analysis (NMR/GCMS) was performed on samples at early (A600 = 0.25), mid-log (0.5-0.7) and late-log phases (1.0-2.0) of growth. Transcriptomic analysis revealed many gene clusters including three novel ABC-type sugar transport clusters to be upregulated in Bi-26 involved in processing of 2'-FL along with metabolism of its monomers glucose, fucose and galactose. Metabolite data confirmed the production of formate, acetate, 1,2-propanediol, lactate and cleaving of fucose from 2'-FL. The formation of acetate, formate, and lactate showed how the cell uses metabolites during fermentation to produce higher levels of ATP (mid-log compared to other stages) or generate cofactors to balance redox. We concluded that 2'-FL metabolism is a complex process involving multiple gene clusters, that produce a more diverse metabolite profile compared to lactose. These results provide valuable insight on the mode-of-action of 2'-FL utilization by Bifidobacterium longum subsp. infantis Bi-26.
Collapse
|
114
|
Enzymatic synthesis of β-galactosyl fucose using recombinant bifidobacterial β-galactosidase and its prebiotic effect. Glycoconj J 2019; 36:199-209. [DOI: 10.1007/s10719-019-09871-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/24/2019] [Accepted: 04/09/2019] [Indexed: 01/05/2023]
|
115
|
Mills S, Stanton C, Lane JA, Smith GJ, Ross RP. Precision Nutrition and the Microbiome, Part I: Current State of the Science. Nutrients 2019; 11:nu11040923. [PMID: 31022973 PMCID: PMC6520976 DOI: 10.3390/nu11040923] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/10/2019] [Accepted: 04/17/2019] [Indexed: 12/11/2022] Open
Abstract
The gut microbiota is a highly complex community which evolves and adapts to its host over a lifetime. It has been described as a virtual organ owing to the myriad of functions it performs, including the production of bioactive metabolites, regulation of immunity, energy homeostasis and protection against pathogens. These activities are dependent on the quantity and quality of the microbiota alongside its metabolic potential, which are dictated by a number of factors, including diet and host genetics. In this regard, the gut microbiome is malleable and varies significantly from host to host. These two features render the gut microbiome a candidate ‘organ’ for the possibility of precision microbiomics—the use of the gut microbiome as a biomarker to predict responsiveness to specific dietary constituents to generate precision diets and interventions for optimal health. With this in mind, this two-part review investigates the current state of the science in terms of the influence of diet and specific dietary components on the gut microbiota and subsequent consequences for health status, along with opportunities to modulate the microbiota for improved health and the potential of the microbiome as a biomarker to predict responsiveness to dietary components. In particular, in Part I, we examine the development of the microbiota from birth and its role in health. We investigate the consequences of poor-quality diet in relation to infection and inflammation and discuss diet-derived microbial metabolites which negatively impact health. We look at the role of diet in shaping the microbiome and the influence of specific dietary components, namely protein, fat and carbohydrates, on gut microbiota composition.
Collapse
Affiliation(s)
- Susan Mills
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland.
| | - Catherine Stanton
- APC Microbiome Ireland, Teagasc Food Research Centre, Fermoy P61 C996, Co Cork, Ireland.
| | - Jonathan A Lane
- H&H Group, Technical Centre, Global Research and Technology Centre, Cork P61 C996, Ireland.
| | - Graeme J Smith
- H&H Group, Technical Centre, Global Research and Technology Centre, Cork P61 C996, Ireland.
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland.
| |
Collapse
|
116
|
Yun EJ, Liu JJ, Lee JW, Kwak S, Yu S, Kim KH, Jin YS. Biosynthetic Routes for Producing Various Fucosyl-Oligosaccharides. ACS Synth Biol 2019; 8:415-424. [PMID: 30668900 DOI: 10.1021/acssynbio.8b00436] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Fucosyl-oligosaccharides (FOSs) play physiologically important roles as prebiotics, neuronal growth factors, and inhibitors of enteropathogens. However, challenges in designed synthesis and mass production of FOSs hamper their industrial applications. Here, we report flexible biosynthetic routes to produce various FOSs, including unnatural ones, through in vitro enzymatic reactions of various sugar acceptors, such as glucose, cellobiose, and agarobiose, and GDP-l-fucose as the fucose donor by using α1,2-fucosyltransferase (FucT2). Also, the whole-cell conversion for fucosylation of various sugar acceptors by overexpressing the genes associated with GDP-l-fucose production and fucT2 gene in Escherichia coli was demonstrated by producing 17.74 g/L of 2'-fucosylgalactose (2'-FG). Prebiotic effects of 2'-FG were verified on the basis of selective fermentability of 2'-FG by probiotic bifidobacteria. These biosynthetic routes can be used to engineer industrial microorganisms for more economical, more flexible, and safer production of FOSs than chemical synthesis of FOSs.
Collapse
Affiliation(s)
- Eun Ju Yun
- Department of Biotechnology, Graduate School, Korea University, Seoul 02841, Republic of Korea
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Jing-Jing Liu
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Jae Won Lee
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Suryang Kwak
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Sora Yu
- Department of Biotechnology, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Kyoung Heon Kim
- Department of Biotechnology, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Youg-Su Jin
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
117
|
Lee JM, Oh SY, Johnston TV, Ku S, Ji GE. Biocatalysis of Fucodian in Undaria pinnatifida Sporophyll Using Bifidobacterium longum RD47 for Production of Prebiotic Fucosylated Oligosaccharide. Mar Drugs 2019; 17:E117. [PMID: 30769784 PMCID: PMC6409798 DOI: 10.3390/md17020117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/04/2019] [Accepted: 02/09/2019] [Indexed: 11/16/2022] Open
Abstract
Fucosylated oligosaccharide (FO) is known to selectively promote the growth of probiotic bacteria and is currently marketed as a functional health food and prebiotic in infant formula. Despite widespread interest in FO among functional food customers, high production costs due to high raw material costs, especially those related to fucose, are a significant production issue. Therefore, several actions are required before efficient large-scale operations can occur, including (i) identification of inexpensive raw materials from which fucosylated oligosaccharides may be produced and (ii) development of production methods to which functional food consumers will not object (e.g., no genetically modified organisms (GMOs)). Undaria pinnatifida, commonly called Miyeok in Korea, is a common edible brown seaweed plentiful on the shores of the Korean peninsula. In particular, the sporophyll of Undaria pinnatifida contains significant levels of l-fucose in the form of fucoidan (a marine sulfated polysaccharide). If the l-fucose present in Undaria pinnatifida sporophyll was capable of being separated and recovered, l-fucose molecules could be covalently joined to other monosaccharides via glycosidic linkages, making this FO manufacturing technology of value in the functional food market. In our previous work, β-galactosidase (EC 3.2.2.23) from Bifidobacterium longum RD47 (B. longum RD47) was found to have transglycosylation activity and produce FO using purified l-fucose and lactose as substrates (reference). In this research, crude fucodian hydrolysates were separated and recovered from edible seaweed (i.e., U. pinnatifida sporophyll). The extracted l-fucose was purified via gel permeation and ion exchange chromatographies and the recovered l-fucose was used to synthesize FO. B. longum RD47 successfully transglycosilated and produced FO using l-fucose derived from Undaria pinnatifida and lactose as substrates. To the best of our knowledge, this is the first report of synthesized FO using Bifidobacterium spp.
Collapse
Affiliation(s)
- Jeong Min Lee
- Department of Food and Nutrition, Research Institute of Human Ecology, Seoul National University, Seoul 08826, Korea.
| | - So Young Oh
- Department of Food and Nutrition, Research Institute of Human Ecology, Seoul National University, Seoul 08826, Korea.
| | - Tony V Johnston
- Fermentation Science Program, School of Agriculture, College of Basic and Applied Sciences, Middle Tennessee State University, Murfreesboro, TN 37132, USA.
| | - Seockmo Ku
- Fermentation Science Program, School of Agriculture, College of Basic and Applied Sciences, Middle Tennessee State University, Murfreesboro, TN 37132, USA.
| | - Geun Eog Ji
- Department of Food and Nutrition, Research Institute of Human Ecology, Seoul National University, Seoul 08826, Korea.
- Research Center, BIFIDO Co., Ltd., Hongcheon 25117, Korea.
| |
Collapse
|
118
|
Fucosylated Human Milk Oligosaccharides and N-Glycans in the Milk of Chinese Mothers Regulate the Gut Microbiome of Their Breast-Fed Infants during Different Lactation Stages. mSystems 2018; 3:mSystems00206-18. [PMID: 30637338 PMCID: PMC6306508 DOI: 10.1128/msystems.00206-18] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/03/2018] [Indexed: 02/07/2023] Open
Abstract
Human milk glycans provide a broad range of carbon sources for gut microbes in infants. Levels of protein glycosylation in human milk vary during lactation and may also be affected by the stages of gestation and lactation and by the secretor status of the mother. This was the first study to evaluate systematically dynamic changes in human milk oligosaccharides and fucosylated N-glycans in the milk of Chinese mothers with different secretor statuses during 6 months of lactation. Given the unique single nucleotide polymorphism site (rs1047781, A385T) on the fucosyltransferase 2 gene among Chinese populations, our report provides a specific insight into the milk glycobiome of Chinese mothers, which may exert effects on the gut microbiota of infants that differ from findings from other study cohorts. The milk glycobiome has a significant impact on the gut microbiota of infants, which plays a pivotal role in health and development. Fucosylated human milk oligosaccharides (HMOs) and N-glycans on milk proteins are beneficial for the development of healthy gut microbiota, and the fucosylation levels of these glycans can be affected by the maternal fucosyltransferase 2 gene (FUT2). Here, we present results of longitudinal research on paired milk and stool samples from 56 Chinese mothers (CMs) and their breast-fed children. Changes of HMOs and fucosylated N-glycans in milk of CMs at different lactation stages were detected, which allowed characterization of the major differences in milk glycans and consequential effects on the gut microbiome of infants according to maternal FUT2 status. Significant differences in the abundance of total and fucosylated HMOs between secretor and nonsecretor CMs were noted, especially during early lactation. Despite a tendency toward decreasing milk protein concentrations, the fucosylation levels of milk N-glycans increased during late lactation. The changes in the levels of fucosylated HMOs and milk N-glycans were highly correlated with the growth of Bifidobacterium spp. and Lactobacillus spp. in the gut of infants during early and later lactation, respectively. Enriched expression of genes encoding glycoside hydrolases, glycosyl transferases, ATP-binding cassette (ABC) transporters, and permeases in infants fed by secretor CMs contributed to the promotion of these bacteria in infants. Our data highlight the important role of fucosylated milk glycans in shaping the gut microbiome of infants and provide a solid foundation for development of “personalized” nutrition for Chinese infants. IMPORTANCE Human milk glycans provide a broad range of carbon sources for gut microbes in infants. Levels of protein glycosylation in human milk vary during lactation and may also be affected by the stages of gestation and lactation and by the secretor status of the mother. This was the first study to evaluate systematically dynamic changes in human milk oligosaccharides and fucosylated N-glycans in the milk of Chinese mothers with different secretor statuses during 6 months of lactation. Given the unique single nucleotide polymorphism site (rs1047781, A385T) on the fucosyltransferase 2 gene among Chinese populations, our report provides a specific insight into the milk glycobiome of Chinese mothers, which may exert effects on the gut microbiota of infants that differ from findings from other study cohorts.
Collapse
|
119
|
Lu J, Claud EC. Connection between gut microbiome and brain development in preterm infants. Dev Psychobiol 2018; 61:739-751. [PMID: 30460694 DOI: 10.1002/dev.21806] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 09/30/2018] [Accepted: 10/17/2018] [Indexed: 12/21/2022]
Abstract
Dysbiosis of the gut microbiome in preterm infants predisposes the neonate to various major morbidities including neonatal necrotizing enterocolitis and sepsis in the neonatal intensive care unit, and adverse neurological outcomes later in life. There are parallel early developmental windows for the gut microbiota and the nervous system during prenatal to postnatal of life. Therefore, preterm infants represent a unique population in which optimization of initial colonization and microbiota development can affect brain development and enhance neurological outcomes. In this review, we will first discuss the factors affecting the assembly of neonatal gut microbiota and the contribution of dysbiosis in preterm infants to neuroinflammation and neurodevelopmental disorders. We then will discuss the emerging pathways connecting the gut microbiome and brain development. Further we will discuss the significance of current models for alteration of the gut microbiome (including humanized gnotobiotic models and exposure to antibiotics) to brain development and functions. Understanding the role of early optimization of the microbiome in brain development is of paramount importance for developing microbiome-targeted therapies and protecting infants from prematurity-related neurodevelopmental diseases.
Collapse
Affiliation(s)
- Jing Lu
- Department of Pediatrics, University of Chicago, Pritzker School of Medicine, Chicago, Illinois
| | - Erika C Claud
- Department of Pediatrics, University of Chicago, Pritzker School of Medicine, Chicago, Illinois
| |
Collapse
|
120
|
Regional variations in human milk oligosaccharides in Vietnam suggest FucTx activity besides FucT2 and FucT3. Sci Rep 2018; 8:16790. [PMID: 30429485 PMCID: PMC6235895 DOI: 10.1038/s41598-018-34882-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 10/18/2018] [Indexed: 12/21/2022] Open
Abstract
Breastfeeding is the normal way of providing young infants with the nutrients they need for healthy growth and development (WHO). Human milk oligosaccharides (hMOS) constitute a highly important class of nutrients that are attracting strong attention in recent years. Several studies have indicated that hMOS have prebiotic properties, but also are effective in anti-adhesion of pathogens, modulating the immune system and providing nutrients for brain growth and development. Most of the latter functions seem to be linked to the presence of fucose-containing immunodeterminant epitopes, and Neu5Ac-bearing oligosaccharides. Analysis of hMOS isolated from 101 mothers’ milk showed regional variation in Lewis- and Secretor based immunodeterminants. Lewis-negative milk groups could be sub-divided into two sub-groups, based on the activity of a third and hitherto unidentified fucosyltransferase enzyme. Analysis of hMOS remaining in faeces showed three sub-groups based on hMOS surviving passage through the gut, full consumption, specific partial consumption and non-specific partial consumption, fitting previous findings.
Collapse
|
121
|
Reverri EJ, Devitt AA, Kajzer JA, Baggs GE, Borschel MW. Review of the Clinical Experiences of Feeding Infants Formula Containing the Human Milk Oligosaccharide 2'-Fucosyllactose. Nutrients 2018; 10:nu10101346. [PMID: 30241407 PMCID: PMC6213476 DOI: 10.3390/nu10101346] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/06/2018] [Accepted: 09/16/2018] [Indexed: 12/14/2022] Open
Abstract
Human milk oligosaccharides (HMOs) are the third most abundant solid component in human milk after lactose and lipids. Preclinical research has demonstrated that HMOs and specifically 2′-fucosyllactose (2′-FL) are more than a prebiotic and have multiple functions, including immune, gut, and cognition benefits. Previously, human milk has been the only source for significant levels of HMOs. The most abundant HMO in most mothers’ breast milk is 2′-FL. Recently, 2′-FL has been synthesized and shown to be structurally identical to the 2′-FL found in human milk. 2′-FL HMO is now available in some commercial infant formulas. The purpose of this narrative review was to summarize the clinical experiences of feeding infant formula supplemented with the HMO, 2′-FL. Most of these studies investigated standard intact milk protein-based infant formulas containing 2′-FL, and one evaluated a partially hydrolyzed whey-based formula. Collectively, these clinical experiences demonstrated that 2′-FL being added to infant formula was safe, well-tolerated, and absorbed and excreted with similar efficiency to 2′-FL in human milk. Further, infants that were fed formula with 2′-FL had immune benefits, fewer parent-reported respiratory infections, and improved symptoms of formula intolerance. Ultimately, infant formula with 2′-FL supports immune and gut health and is closer compositionally and functionally to human milk.
Collapse
Affiliation(s)
| | - Amy A Devitt
- Abbott Nutrition, Abbott Laboratories, Columbus, OH 43219, USA.
| | - Janice A Kajzer
- Abbott Nutrition, Abbott Laboratories, Columbus, OH 43219, USA.
| | | | | |
Collapse
|
122
|
Rajani PS, Seppo AE, Järvinen KM. Immunologically Active Components in Human Milk and Development of Atopic Disease, With Emphasis on Food Allergy, in the Pediatric Population. Front Pediatr 2018; 6:218. [PMID: 30131949 PMCID: PMC6090044 DOI: 10.3389/fped.2018.00218] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/16/2018] [Indexed: 12/17/2022] Open
Abstract
Breast-feeding is currently recommended to prevent the development of allergic diseases; however, data are conflicting and mechanisms are unclear. The immunomodulatory composition of human milk is poorly characterized and varies between mothers. We and others have shown that high levels of human milk IgA and certain cytokines and human milk oligosaccharides are associated with protection against food allergy in the infant, but it is unclear whether they are responsible for or simply biomarkers of the vertical transfer of protection. Because human milk has pre- and probiotic properties, the anti-allergy protection afforded by human milk may be due to its control on the developing gut microbiome. In mice, murine milk IgA supports gut homeostasis and shapes the microbiota, which in turn diversifies the intestinal IgA repertoire that reciprocally promotes the diversity of gut microbiome; these mechanisms are poorly understood in humans. In addition, several human milk bioactives are immunostimulatory, which may in part provide protection against allergic diseases. The regulation of immunologically active components in human milk is incompletely understood, although accumulating evidence suggests that IgA and cytokines in human milk reflect maternal exposures. This review summarizes the current literature on human milk components that have been associated with protection against food allergy and related allergic disorders in early childhood and discusses the work relating to regulation of these levels in human milk and possible mechanisms of action.
Collapse
Affiliation(s)
| | | | - Kirsi M. Järvinen
- Division of Pediatric Allergy and Immunology and Center for Food Allergy, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| |
Collapse
|
123
|
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
|
124
|
Lindberg TP, Caimano MJ, Hagadorn JI, Bennett EM, Maas K, Brownell EA, Matson AP. Preterm infant gut microbial patterns related to the development of necrotizing enterocolitis. J Matern Fetal Neonatal Med 2018; 33:349-358. [PMID: 29909714 DOI: 10.1080/14767058.2018.1490719] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objectives: To define gut microbial patterns in preterm infants with and without necrotizing enterocolitis (NEC) and to characterize clinical factors related to the composition of the preterm intestinal microbiome.Methods: Fecal samples were collected at one-week intervals from infants with gestational ages <30 weeks at a single level IV neonatal intensive care unit. Using 16S rRNA gene sequencing, the composition and diversity of microbiota were determined in samples collected from five NEC infants and five matched controls. Hierarchical linear regression was used to identify clinical factors related to microbial diversity and specific bacterial signatures.Results: Low levels of diversity were demonstrated in samples obtained from all preterm infants and antibiotic exposure further decreased diversity among both NEC cases and controls. Fecal microbial composition differed between NEC cases and controls, with a greater abundance of Proteobacteria and bacteria belonging to the class Gammaproteobacteria among NEC infants. Control infants demonstrated a greater abundance of bacteria belonging to the phylum Firmicutes.Conclusion: These findings indicate that an association exists between intestinal Proteobacteria and NEC, and strengthens the notion that an overly exuberant response to Gram-negative products, particularly lipopolysaccharide, in the preterm intestine is involved in NEC pathogenesis. Cumulative exposure to antibiotics corresponded to a reduction in microbial diversity in both NEC cases and controls.
Collapse
Affiliation(s)
- Tristan P Lindberg
- Division of Neonatology, Connecticut Children's Medical Center, Hartford, CT, USA.,University of Connecticut School of Medicine, Farmington, CT, USA
| | | | - James I Hagadorn
- Division of Neonatology, Connecticut Children's Medical Center, Hartford, CT, USA.,University of Connecticut School of Medicine, Farmington, CT, USA
| | - Erin M Bennett
- Division of Research, Connecticut Children's Medical Center, Hartford, CT, USA
| | | | - Elizabeth A Brownell
- Division of Neonatology, Connecticut Children's Medical Center, Hartford, CT, USA.,University of Connecticut School of Medicine, Farmington, CT, USA.,Division of Research, Connecticut Children's Medical Center, Hartford, CT, USA
| | - Adam P Matson
- Division of Neonatology, Connecticut Children's Medical Center, Hartford, CT, USA.,University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
125
|
Yu J, Shin J, Park M, Seydametova E, Jung SM, Seo JH, Kweon DH. Engineering of α-1,3-fucosyltransferases for production of 3-fucosyllactose in Escherichia coli. Metab Eng 2018; 48:269-278. [DOI: 10.1016/j.ymben.2018.05.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/12/2018] [Accepted: 05/31/2018] [Indexed: 12/23/2022]
|
126
|
Durack J, Kimes NE, Lin DL, Rauch M, McKean M, McCauley K, Panzer AR, Mar JS, Cabana MD, Lynch SV. Delayed gut microbiota development in high-risk for asthma infants is temporarily modifiable by Lactobacillus supplementation. Nat Commun 2018; 9:707. [PMID: 29453431 PMCID: PMC5816017 DOI: 10.1038/s41467-018-03157-4] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/24/2018] [Indexed: 12/11/2022] Open
Abstract
Gut microbiota dysbiosis and metabolic dysfunction in infancy precedes childhood atopy and asthma development. Here we examined gut microbiota maturation over the first year of life in infants at high risk for asthma (HR), and whether it is modifiable by early-life Lactobacillus supplementation. We performed a longitudinal comparison of stool samples collected from HR infants randomized to daily oral Lactobacillus rhamnosus GG (HRLGG) or placebo (HRP) for 6 months, and healthy (HC) infants. Meconium microbiota of HRP participants is distinct, follows a delayed developmental trajectory, and is primarily glycolytic and depleted of a range of anti-inflammatory lipids at 6 months of age. These deficits are partly rescued in HRLGG infants, but this effect was lost at 12 months of age, 6 months after cessation of supplementation. Thus we show that early-life gut microbial development is distinct, but plastic, in HR infants. Our findings offer a novel strategy for early-life preventative interventions. Gut microbial dysbiosis in infancy is associated with childhood atopy and the development of asthma. Here, the authors show that gut microbiota perturbation is evident in the very earliest stages of postnatal life, continues throughout infancy, and can be partially rescued by Lactobacillus supplementation in high-risk for asthma infants.
Collapse
Affiliation(s)
- Juliana Durack
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Nikole E Kimes
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA.,Siolta Therapeutics, 953 Indiana Street, San Francisco, CA, 94107, USA
| | - Din L Lin
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Marcus Rauch
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA.,Janssen Prevention Center, 2 Royal College Street, London, NW1 0NH, UK
| | - Michelle McKean
- Division of General Pediatrics, Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Kathryn McCauley
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ariane R Panzer
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jordan S Mar
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA.,Genentech, 340 Pt. San Bruno Boulevard, South San Francisco, CA, 94080, USA
| | - Michael D Cabana
- Division of General Pediatrics, Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA.,Division of Clinical Epidemiology, Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Susan V Lynch
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
127
|
Abstract
NEC is a multifactorial disease that occurs when multiple risk factors and/or stressors overlap, leading to profound inflammation and intestinal injury. Human milk feedings, both from the infant's mother and donor human milk, have been associated with reductions in NEC in preterm infants. This article will review the protective factors in human milk, clinical studies of human milk and NEC, and practices to enhance human milk use in neonatal intensive care units.
Collapse
Affiliation(s)
- Aloka L Patel
- Section of Neonatology, Rush University Children's Hospital, 1653 W. Congress Pkwy, Pavilion 353, Chicago, Illinois 60612.
| | - Jae H Kim
- Divisions of Neonatology & Pediatric Gastroenterology, Hepatology and Nutrition, University of California San Diego, Rady Children's Hospital of San Diego, San Diego, California
| |
Collapse
|
128
|
Doherty AM, Lodge CJ, Dharmage SC, Dai X, Bode L, Lowe AJ. Human Milk Oligosaccharides and Associations With Immune-Mediated Disease and Infection in Childhood: A Systematic Review. Front Pediatr 2018; 6:91. [PMID: 29732363 PMCID: PMC5920034 DOI: 10.3389/fped.2018.00091] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/23/2018] [Indexed: 12/27/2022] Open
Abstract
Complex sugars found in breastmilk, human milk oligosaccharides (HMOs), may assist in early-life immune programming and prevention against infectious diseases. This study aimed to systematically review the associations between maternal levels of HMOs and development of immune-mediated or infectious diseases in the offspring. PubMed and EMBASE databases were searched (last search on 22 February 2018) according to a predetermined search strategy. Original studies published in English examining the effect of HMOs on immune-mediated and infectious disease were eligible for inclusion. Of 847 identified records, 10 articles from 6 original studies were included, with study quality ranging from low to high. Of three studies to examine allergic disease outcomes, one reported a protective effect against cow's milk allergy (CMA) by 18 months of age associated with lower lacto-N-fucopentaose (LNFP) III concentrations (OR: 6.7, 95% CI 2.0-22). Another study found higher relative abundance of fucosyloligosaccharides was associated with reduced diarrhea incidence by 2 years, due to (i) stable toxin-E. coli infection (p = 0.04) and (ii) "all causes" (p = 0.042). Higher LNFP-II concentrations were associated with (i) reduced cases of gastroenteritis and respiratory tract infections at 6 weeks (p = 0.004, p = 0.010) and 12 weeks (p = 0.038, p = 0.038) and (ii) reduced HIV transmission (OR: 0.45; 95% CI: 0.21-0.97) and mortality risk among HIV-exposed, uninfected infants (HR: 0.33; 95% CI: 0.14-0.74) by 24 months. Due to heterogeneity of the outcomes reported, pooling of results was not possible. There was limited evidence that low concentrations of LNFP-III are associated with CMA and that higher fucosyloligosaccharide levels protect infants against infectious disease. Further research is needed.
Collapse
Affiliation(s)
- Alice M Doherty
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Caroline J Lodge
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia.,Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Shyamali C Dharmage
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia.,Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Xin Dai
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Lars Bode
- Division of Neonatology, Department of Pediatrics and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (LRF MoMI CoRE), University of California, San Diego, La Jolla, CA, United States.,Division of Pediatric Gastroenterology and Nutrition, Department of Pediatrics and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (LRF MoMI CoRE), University of California, San Diego, La Jolla, CA, United States
| | - Adrian J Lowe
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia.,Murdoch Children's Research Institute, Melbourne, VIC, Australia
| |
Collapse
|
129
|
Ayechu-Muruzabal V, van Stigt AH, Mank M, Willemsen LEM, Stahl B, Garssen J, Van't Land B. Diversity of Human Milk Oligosaccharides and Effects on Early Life Immune Development. Front Pediatr 2018; 6:239. [PMID: 30250836 PMCID: PMC6140589 DOI: 10.3389/fped.2018.00239] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/07/2018] [Indexed: 12/12/2022] Open
Abstract
One of the well-known features of human milk, is the capacity to protect against the risk and impact of neonatal infections, as well as to influence the onset of allergic and metabolic disease manifestations. The major objective of this review is to provide a detailed overview regarding the role of human milk, more specifically the diversity in human milk oligosaccharides (HMOS), on early life immune development. Novel insights in immune modulatory effects of HMOS obtained by in vitro as well as in vivo studies, adds to the understanding on how early life nutrition may impact immune development. Extensive description and analysis of single HMOS contributing to the diversity within the composition provided during breastfeeding will be discussed with specific emphasis on immune development and the susceptibility to neonatal and childhood infections.
Collapse
Affiliation(s)
- Veronica Ayechu-Muruzabal
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Arthur H van Stigt
- Pediatric Immunology, Laboratory of Translational Immunology, The Wilhelmina Children's Hospital, University Medical Center, Utrecht, Netherlands
| | - Marko Mank
- Department of Immunology and Department of Human Milk Research & Analytical Science, Danone Nutricia Research, Utrecht, Netherlands
| | - Linette E M Willemsen
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Bernd Stahl
- Department of Immunology and Department of Human Milk Research & Analytical Science, Danone Nutricia Research, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands.,Department of Immunology and Department of Human Milk Research & Analytical Science, Danone Nutricia Research, Utrecht, Netherlands
| | - Belinda Van't Land
- Pediatric Immunology, Laboratory of Translational Immunology, The Wilhelmina Children's Hospital, University Medical Center, Utrecht, Netherlands.,Department of Immunology and Department of Human Milk Research & Analytical Science, Danone Nutricia Research, Utrecht, Netherlands
| |
Collapse
|
130
|
Yebra MJ, Monedero V, Rodríguez-Díaz J, Bidart GN, Becerra JE. Bioactive Properties and Biotechnological Production of Human Milk Oligosaccharides. ADVANCES IN BIOTECHNOLOGY FOR FOOD INDUSTRY 2018:425-460. [DOI: 10.1016/b978-0-12-811443-8.00015-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
131
|
Xiao L, Van't Land B, van de Worp WRPH, Stahl B, Folkerts G, Garssen J. Early-Life Nutritional Factors and Mucosal Immunity in the Development of Autoimmune Diabetes. Front Immunol 2017; 8:1219. [PMID: 29033938 PMCID: PMC5626949 DOI: 10.3389/fimmu.2017.01219] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 09/14/2017] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes (T1D) is an immune-mediated disease with a strong genetic basis but might be influenced by non-genetic factors such as microbiome development that “programs” the immune system during early life as well. Factors influencing pathogenesis, including a leaky intestinal mucosal barrier, an aberrant gut microbiota composition, and altered immune responsiveness, offer potential targets for prevention and/or treatment of T1D through nutritional or pharmacologic means. In this review, nutritional approaches during early life in order to protect against T1D development have been discussed. The critical role of tolerogenic dendritic cells in central and peripheral tolerance has been emphasized. In addition, since the gut microbiota affects the development of T1D through short-chain fatty acid (SCFA)-dependent mechanisms, we hypothesize that nutritional intervention boosting SCFA production may be used as a novel prevention strategy. Current retrospective evidence has suggested that exclusive and prolonged breastfeeding might play a protective role against the development of T1D. The beneficial properties of human milk are possibly attributed to its bioactive components such as unique immune-modulatory components human milk oligosaccharides and metabolites derived thereof, including SCFAs. These components might play a key role in healthy immune development and creating a fit and resilient immune system in early and later life.
Collapse
Affiliation(s)
- Ling Xiao
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Belinda Van't Land
- Nutricia Research, Utrecht, Netherlands.,Department of Pediatric Immunology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Wouter R P H van de Worp
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | | | - Gert Folkerts
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Nutricia Research, Utrecht, Netherlands
| |
Collapse
|
132
|
Sprenger GA, Baumgärtner F, Albermann C. Production of human milk oligosaccharides by enzymatic and whole-cell microbial biotransformations. J Biotechnol 2017; 258:79-91. [PMID: 28764968 DOI: 10.1016/j.jbiotec.2017.07.030] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 12/14/2022]
Abstract
Human milk oligosaccharides (HMO) are almost unique constituents of breast milk and are not found in appreciable amounts in cow milk. Due to several positive aspects of HMO for the development, health, and wellbeing of infants, production of HMO would be desirable. As a result, scientists from different disciplines have developed methods for the preparation of single HMO compounds. Here, we review approaches to HMO preparation by (chemo-)enzymatic syntheses or by whole-cell biotransformation with recombinant bacterial cells. With lactose as acceptor (in vitro or in vivo), fucosyltransferases can be used for the production of 2'-fucosyllactose, 3-fucosyllactose, or more complex fucosylated core structures. Sialylated HMO can be produced by sialyltransferases and trans-sialidases. Core structures as lacto-N-tetraose can be obtained by glycosyltransferases from chemical donor compounds or by multi-enzyme cascades; recent publications also show production of lacto-N-tetraose by recombinant Escherichia coli bacteria and approaches to obtain fucosylated core structures. In view of an industrial production of HMOs, the whole cell biotransformation is at this stage the most promising option to provide human milk oligosaccharides as food additive.
Collapse
Affiliation(s)
- Georg A Sprenger
- Institute of Microbiology, University of Stuttgart, Allmandring 31, D-70569 Stuttgart, Germany.
| | - Florian Baumgärtner
- Institute of Microbiology, University of Stuttgart, Allmandring 31, D-70569 Stuttgart, Germany
| | - Christoph Albermann
- Institute of Microbiology, University of Stuttgart, Allmandring 31, D-70569 Stuttgart, Germany
| |
Collapse
|
133
|
Laforest-Lapointe I, Arrieta MC. Patterns of Early-Life Gut Microbial Colonization during Human Immune Development: An Ecological Perspective. Front Immunol 2017; 8:788. [PMID: 28740492 PMCID: PMC5502328 DOI: 10.3389/fimmu.2017.00788] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 06/22/2017] [Indexed: 12/24/2022] Open
Abstract
Alterations in gut microbial colonization during early life have been reported in infants that later developed asthma, allergies, type 1 diabetes, as well as in inflammatory bowel disease patients, previous to disease flares. Mechanistic studies in animal models have established that microbial alterations influence disease pathogenesis via changes in immune system maturation. Strong evidence points to the presence of a window of opportunity in early life, during which changes in gut microbial colonization can result in immune dysregulation that predisposes susceptible hosts to disease. Although the ecological patterns of microbial succession in the first year of life have been partly defined in specific human cohorts, the taxonomic and functional features, and diversity thresholds that characterize these microbial alterations are, for the most part, unknown. In this review, we summarize the most important links between the temporal mosaics of gut microbial colonization and the age-dependent immune functions that rely on them. We also highlight the importance of applying ecology theory to design studies that explore the interactions between this complex ecosystem and the host immune system. Focusing research efforts on understanding the importance of temporally structured patterns of diversity, keystone groups, and inter-kingdom microbial interactions for ecosystem functions has great potential to enable the development of biologically sound interventions aimed at maintaining and/or improving immune system development and preventing disease.
Collapse
Affiliation(s)
- Isabelle Laforest-Lapointe
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
| | - Marie-Claire Arrieta
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
134
|
Thursby E, Juge N. Introduction to the human gut microbiota. Biochem J 2017; 474:1823-1836. [PMID: 28512250 PMCID: PMC5433529 DOI: 10.1042/bcj20160510] [Citation(s) in RCA: 2010] [Impact Index Per Article: 251.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 02/08/2023]
Abstract
The human gastrointestinal (GI) tract harbours a complex and dynamic population of microorganisms, the gut microbiota, which exert a marked influence on the host during homeostasis and disease. Multiple factors contribute to the establishment of the human gut microbiota during infancy. Diet is considered as one of the main drivers in shaping the gut microbiota across the life time. Intestinal bacteria play a crucial role in maintaining immune and metabolic homeostasis and protecting against pathogens. Altered gut bacterial composition (dysbiosis) has been associated with the pathogenesis of many inflammatory diseases and infections. The interpretation of these studies relies on a better understanding of inter-individual variations, heterogeneity of bacterial communities along and across the GI tract, functional redundancy and the need to distinguish cause from effect in states of dysbiosis. This review summarises our current understanding of the development and composition of the human GI microbiota, and its impact on gut integrity and host health, underlying the need for mechanistic studies focusing on host-microbe interactions.
Collapse
Affiliation(s)
- Elizabeth Thursby
- The Gut Health and Food Safety Programme, Institute of Food Research, Norwich Research Park, Norwich NR4 7UA, U.K
| | - Nathalie Juge
- The Gut Health and Food Safety Programme, Institute of Food Research, Norwich Research Park, Norwich NR4 7UA, U.K.
| |
Collapse
|
135
|
Haque SZ, Haque M. The ecological community of commensal, symbiotic, and pathogenic gastrointestinal microorganisms - an appraisal. Clin Exp Gastroenterol 2017; 10:91-103. [PMID: 28503071 PMCID: PMC5426469 DOI: 10.2147/ceg.s126243] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The human gastrointestinal tract is inhabited by a vast population of bacteria, numbering ~100 trillion. These microorganisms have been shown to play a significant role in digestion, metabolism, and the immune system. The aim of this study was to review and discuss how the human body interacts with its gut microbiome and in turn the effects that the microorganisms have on its host, overall resulting in a true mutualistic relationship.
Collapse
Affiliation(s)
- Seraj Zohurul Haque
- School of Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, Scotland, UK
| | - Mainul Haque
- Unit of Pharmacology, Faculty of Medicine and Defense Health, National Defense University of Malaysia, Kem Sungai Besi, Kuala Lumpur, Malaysia
| |
Collapse
|
136
|
Martín-Ortiz A, Barile D, Salcedo J, Moreno FJ, Clemente A, Ruiz-Matute AI, Sanz ML. Changes in Caprine Milk Oligosaccharides at Different Lactation Stages Analyzed by High Performance Liquid Chromatography Coupled to Mass Spectrometry. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:3523-3531. [PMID: 28393524 PMCID: PMC5557284 DOI: 10.1021/acs.jafc.6b05104] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Changes of the abundance of caprine milk oligosaccharides (CMO) at different lactation stages have been evaluated by hydrophilic interaction liquid chromatography coupled to mass spectrometry (HILIC-Q MS) and nanoflow liquid chromatography-quadrupole-time-of-flight mass spectrometry (nano-LC-Chip-QTOF MS). Eight major oligosaccharides (OS) were quantified at different lactation stages by HILIC-Q MS, while the use of nano-LC-Chip-QToF MS allowed expanding the study to forty-nine different OS by monitoring neutral non- and fucosylated species, as well as acidic species containing not only N-acetyl-neuraminic acid or N-glycolyl-neuraminic acid residues but also the combination of both sialic acids. Overall, the most abundant OS decreased with lactation time, whereas different trends were observed for minor OS. 6'-Sialyl-lactose was the most abundant acidic OS while galactosyl-lactose isomers were identified as the most abundant neutral OS. This is the first time that a comprehensive study regarding the changes of the abundance of CMO, both neutral and acidic, at different lactation stages is carried out.
Collapse
Affiliation(s)
- Andrea Martín-Ortiz
- Instituto de Química Orgánica General (CSIC), Juan de la Cierva, 3, 28006 Madrid, Spain
| | - Daniela Barile
- Department of Food Science and Technology, University of California Davis, One Shields Avenue, Davis, California 95616, United States
| | - Jaime Salcedo
- Department of Food Science and Technology, University of California Davis, One Shields Avenue, Davis, California 95616, United States
| | - F. Javier Moreno
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), C/Nicolás Cabrera, 9, Campus de Cantoblanco -Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Alfonso Clemente
- Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas (CSIC), Profesor Albareda 1, 18008 Granada, Spain
| | - Ana I. Ruiz-Matute
- Instituto de Química Orgánica General (CSIC), Juan de la Cierva, 3, 28006 Madrid, Spain
- Corresponding Author:. Tel. +34915622900. Fax: +34915644853
| | - María L. Sanz
- Instituto de Química Orgánica General (CSIC), Juan de la Cierva, 3, 28006 Madrid, Spain
| |
Collapse
|
137
|
Wampach L, Heintz-Buschart A, Hogan A, Muller EEL, Narayanasamy S, Laczny CC, Hugerth LW, Bindl L, Bottu J, Andersson AF, de Beaufort C, Wilmes P. Colonization and Succession within the Human Gut Microbiome by Archaea, Bacteria, and Microeukaryotes during the First Year of Life. Front Microbiol 2017; 8:738. [PMID: 28512451 PMCID: PMC5411419 DOI: 10.3389/fmicb.2017.00738] [Citation(s) in RCA: 191] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 04/10/2017] [Indexed: 12/26/2022] Open
Abstract
Perturbations to the colonization process of the human gastrointestinal tract have been suggested to result in adverse health effects later in life. Although much research has been performed on bacterial colonization and succession, much less is known about the other two domains of life, archaea, and eukaryotes. Here we describe colonization and succession by bacteria, archaea and microeukaryotes during the first year of life (samples collected around days 1, 3, 5, 28, 150, and 365) within the gastrointestinal tract of infants delivered either vaginally or by cesarean section and using a combination of quantitative real-time PCR as well as 16S and 18S rRNA gene amplicon sequencing. Sequences from organisms belonging to all three domains of life were detectable in all of the collected meconium samples. The microeukaryotic community composition fluctuated strongly over time and early diversification was delayed in infants receiving formula milk. Cesarean section-delivered (CSD) infants experienced a delay in colonization and succession, which was observed for all three domains of life. Shifts in prokaryotic succession in CSD infants compared to vaginally delivered (VD) infants were apparent as early as days 3 and 5, which were characterized by increased relative abundances of the genera Streptococcus and Staphylococcus, and a decrease in relative abundance for the genera Bifidobacterium and Bacteroides. Generally, a depletion in Bacteroidetes was detected as early as day 5 postpartum in CSD infants, causing a significantly increased Firmicutes/Bacteroidetes ratio between days 5 and 150 when compared to VD infants. Although the delivery mode appeared to have the strongest influence on differences between the infants, other factors such as a younger gestational age or maternal antibiotics intake likely contributed to the observed patterns as well. Our findings complement previous observations of a delay in colonization and succession of CSD infants, which affects not only bacteria but also archaea and microeukaryotes. This further highlights the need for resolving bacterial, archaeal, and microeukaryotic dynamics in future longitudinal studies of microbial colonization and succession within the neonatal gastrointestinal tract.
Collapse
Affiliation(s)
- Linda Wampach
- Luxembourg Centre for Systems Biomedicine, University of LuxembourgEsch-sur-Alzette, Luxembourg
| | - Anna Heintz-Buschart
- Luxembourg Centre for Systems Biomedicine, University of LuxembourgEsch-sur-Alzette, Luxembourg
| | - Angela Hogan
- Integrated BioBank of LuxembourgLuxembourg, Luxembourg
| | - Emilie E L Muller
- Luxembourg Centre for Systems Biomedicine, University of LuxembourgEsch-sur-Alzette, Luxembourg
| | - Shaman Narayanasamy
- Luxembourg Centre for Systems Biomedicine, University of LuxembourgEsch-sur-Alzette, Luxembourg
| | - Cedric C Laczny
- Luxembourg Centre for Systems Biomedicine, University of LuxembourgEsch-sur-Alzette, Luxembourg
| | - Luisa W Hugerth
- Science for Life Laboratory, Division of Gene Technology, School of Biotechnology, Royal Institute of TechnologyStockholm, Sweden
| | - Lutz Bindl
- Centre Hospitalier de LuxembourgLuxembourg, Luxembourg
| | - Jean Bottu
- Centre Hospitalier de LuxembourgLuxembourg, Luxembourg
| | - Anders F Andersson
- Science for Life Laboratory, Division of Gene Technology, School of Biotechnology, Royal Institute of TechnologyStockholm, Sweden
| | - Carine de Beaufort
- Luxembourg Centre for Systems Biomedicine, University of LuxembourgEsch-sur-Alzette, Luxembourg.,Centre Hospitalier de LuxembourgLuxembourg, Luxembourg
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of LuxembourgEsch-sur-Alzette, Luxembourg
| |
Collapse
|
138
|
Huang D, Yang K, Liu J, Xu Y, Wang Y, Wang R, Liu B, Feng L. Metabolic engineering of Escherichia coli for the production of 2′-fucosyllactose and 3-fucosyllactose through modular pathway enhancement. Metab Eng 2017; 41:23-38. [DOI: 10.1016/j.ymben.2017.03.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/14/2017] [Accepted: 03/08/2017] [Indexed: 01/20/2023]
|
139
|
Gomez DE, Arroyo LG, Costa MC, Viel L, Weese JS. Characterization of the Fecal Bacterial Microbiota of Healthy and Diarrheic Dairy Calves. J Vet Intern Med 2017; 31:928-939. [PMID: 28390070 PMCID: PMC5435056 DOI: 10.1111/jvim.14695] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/13/2017] [Accepted: 02/21/2017] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Neonatal diarrhea accounts for more than 50% of total deaths in dairy calves. Few population-based studies of cattle have investigated how the microbiota is impacted during diarrhea. OBJECTIVES To characterize the fecal microbiota and predict the functional potential of the microbial communities in healthy and diarrheic calves. METHODS Fifteen diarrheic calves between the ages of 1 and 30 days and 15 age-matched healthy control calves were enrolled from 2 dairy farms. The Illumina MiSeq sequencer was used for high-throughput sequencing of the V4 region of the 16S rRNA gene (Illumina, San Diego, CA). RESULTS Significant differences in community membership and structure were identified among healthy calves from different farms. Differences in community membership and structure also were identified between healthy and diarrheic calves within each farm. Based on linear discriminant analysis effect size (LEfSe), the genera Bifidobacterium, Megamonas, and a genus of the family Bifidobacteriaceae were associated with health at farm 1, whereas Lachnospiraceae incertae sedis, Dietzia and an unclassified genus of the family Veillonellaceae were significantly associated with health at farm 2. The Phylogenetic Investigation of Communities Reconstruction of Unobserved States (PICRUSt) analysis indicated that diarrheic calves had decreased abundances of genes responsible for metabolism of various vitamins, amino acids, and carbohydrate. CLINICAL RELEVANCE The fecal microbiota of healthy dairy calves appeared to be farm specific as were the changes observed during diarrhea. The differences in microbiota structure and membership between healthy and diarrheic calves suggest that dysbiosis can occur in diarrheic calves and it is associated with changes in predictive metagenomic function.
Collapse
Affiliation(s)
- D E Gomez
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - L G Arroyo
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - M C Costa
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - L Viel
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - J S Weese
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
140
|
Sprenger N, Odenwald H, Kukkonen AK, Kuitunen M, Savilahti E, Kunz C. FUT2-dependent breast milk oligosaccharides and allergy at 2 and 5 years of age in infants with high hereditary allergy risk. Eur J Nutr 2017; 56:1293-1301. [PMID: 26907090 DOI: 10.1007/s00394-016-1180-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 02/06/2016] [Indexed: 01/19/2023]
Abstract
PURPOSE Manifestation of allergic disease depends on genetic predisposition, diet and commensal microbiota. Genetic polymorphism of mothers determines their breast milk glycan composition. One major determinant is the fucosyltransferase 2 (FUT2, secretor gene) that was shown to be linked to commensal microbiota establishment. We studied whether FUT2-dependent breast milk oligosaccharides are associated with allergic disease in breast-fed infants later in life. METHODS We analyzed FUT2-dependent oligosaccharides in breast milk samples of mothers (n = 266) from the placebo group of a randomized placebo-controlled trial of prebiotics and probiotics as preventive against allergic disease in infants with high allergy risk (trial registry number: NCT00298337). Using logistic regression models, we studied associations between FUT2-dependent breast milk oligosaccharides and incidence of allergic disease at 2 and 5 years of age. RESULTS At 2 years, but not at 5 years of age, we observed a presumed lower incidence (p < 0.1) for IgE-associated eczema manifestation in C-section-born infants who were fed breast milk containing FUT2-dependent oligosaccharides. By logistic regression, we observed a similar relation (p < 0.1) between presence of FUT2-dependent breast milk oligosaccharides and IgE-associated disease and IgE-associated eczema in C-section-born infants only. When testing with the levels of breast milk oligosaccharide 2'-fucosyllactose as proxy for FUT2 activity, we observed significant (p < 0.05) associations in the C-section-born infants with 'any allergic disease,' IgE-associated disease, eczema and IgE-associated eczema. CONCLUSION The data indicate that infants born by C-section and having a high hereditary risk for allergies might have a lower risk to manifest IgE-associated eczema at 2 years, but not 5 years of age, when fed breast milk with FUT2-dependent milk oligosaccharides. Further studies with larger cohorts and especially randomized controlled intervention trials are required to build on these preliminary observations.
Collapse
Affiliation(s)
- Norbert Sprenger
- Nestlé Research Center, Nestec S.A., Vers-Chez-Les-Blanc, 1000, Lausanne 26, Switzerland.
| | - Hannah Odenwald
- Nestlé Research Center, Nestec S.A., Vers-Chez-Les-Blanc, 1000, Lausanne 26, Switzerland
- Institute of Nutritional Sciences, University of Giessen, Wilhelmstr. 20, 35392, Giessen, Germany
- Nestle Health Sciences, Biopole, 1066, Epalinges, Switzerland
| | - Anna Kaarina Kukkonen
- Skin and Allergy Hospital, Helsinki University Central Hospital, 00029, Helsinki, Finland
| | - Mikael Kuitunen
- Children's Hospital, University of Helsinki and Helsinki University Central Hospital, 00029, Helsinki, Finland
| | - Erkki Savilahti
- Children's Hospital, University of Helsinki and Helsinki University Central Hospital, 00029, Helsinki, Finland
| | - Clemens Kunz
- Institute of Nutritional Sciences, University of Giessen, Wilhelmstr. 20, 35392, Giessen, Germany.
| |
Collapse
|
141
|
Nemati S, Teimourian S. An Overview of Inflammatory Bowel Disease: General Consideration and Genetic Screening Approach in Diagnosis of Early Onset Subsets. Middle East J Dig Dis 2017. [PMID: 28638582 PMCID: PMC5471105 DOI: 10.15171/mejdd.2017.54] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Inflammatory bowel disease (IBD) is the consequence of an aberrant hemostasis of the immune cells at the gut mucosal border. Based on clinical manifestation, laboratory tests, radiological studies, endoscopic and histological features, this disease is divided into three main types including Crohn's disease (CD), Ulcerative colitis (UC), and IBDunclassified (IBD-U). IBD is frequently presented in adults, but about 20% of IBD cases are diagnosed during childhood called pediatric IBD (PIBD). Some patients in the latter group emerge the first symptoms during infancy or under 5 years of age named infantile and very early onset IBD (VEO-IBD), respectively. These subtypes make a small fraction of PIBD, but they have exclusive phenotypic and genetic characteristics such that they are accompanied by severe disease course and resistance to conventional therapy. In this context, understanding the underlying molecular pathology opens a promising field for individualized and effective treatment. Here, we describe current hypotheses on IBD pathophysiology then explain the new idea about genetic screening technology as a good potential approach to identify the causal variants early in the disease manifestation, which is especially important for the fast and accurate treatment of VEO-IBD.
Collapse
Affiliation(s)
- Shahram Nemati
- Department of Medical Genetics, Tehran University of Medical Sciences,International Campus (TUMS-IC), Tehran, Iran
| | - Shahram Teimourian
- Department of Medical Genetics, Iran University of Medical Sciences,Tehran, Iran
| |
Collapse
|
142
|
The Fecal Microbial Community of Breast-fed Infants from Armenia and Georgia. Sci Rep 2017; 7:40932. [PMID: 28150690 PMCID: PMC5288704 DOI: 10.1038/srep40932] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 12/12/2016] [Indexed: 01/11/2023] Open
Abstract
Multiple factors help shape the infant intestinal microbiota early in life. Environmental conditions such as the presence of bioactive molecules from breast milk dictate gut microbial growth and survival. Infants also receive distinct, personalized, bacterial exposures leading to differential colonization. Microbial exposures and gut environmental conditions differ between infants in different locations, as does the typical microbial community structure in an infant’s gut. Here we evaluate potential influences on the infant gut microbiota through a longitudinal study on cohorts of breast-fed infants from the neighboring countries of Armenia and Georgia, an area of the world for which the infant microbiome has not been previously investigated. Marker gene sequencing of 16S ribosomal genes revealed that the gut microbial communities of infants from these countries were dominated by bifidobacteria, were different from each other, and were marginally influenced by their mother’s secretor status. Species-level differences in the bifidobacterial communities of each country and birth method were also observed. These community differences suggest that environmental variation between individuals in different locations may influence the gut microbiota of infants.
Collapse
|
143
|
Ackerman DL, Craft KM, Townsend SD. Infant food applications of complex carbohydrates: Structure, synthesis, and function. Carbohydr Res 2017; 437:16-27. [PMID: 27883906 PMCID: PMC6172010 DOI: 10.1016/j.carres.2016.11.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/31/2016] [Accepted: 11/09/2016] [Indexed: 01/05/2023]
Abstract
Professional health bodies such as the World Health Organization (WHO), the American Academy of Pediatrics (AAP), and the U.S. Department of Health and Human Services (HHS) recommend breast milk as the sole source of food during the first year of life. This position recognizes human milk as being uniquely suited for infant nutrition. Nonetheless, most neonates in the West are fed alternatives by 6 months of age. Although inferior to human milk in most aspects, infant formulas are able to promote effective growth and development. However, while breast-fed infants feature a microbiota dominated by bifidobacteria, the bacterial flora of formula-fed infants is usually heterogeneous with comparatively lower levels of bifidobacteria. Thus, the objective of any infant food manufacturer is to prepare a product that results in a formula-fed infant developing a breast-fed infant-like microbiota. The goal of this focused review is to discuss the structure, synthesis, and function of carbohydrate additives that play a role in governing the composition of the infant microbiome and have other health benefits.
Collapse
Affiliation(s)
- Dorothy L Ackerman
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, United States
| | - Kelly M Craft
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, United States
| | - Steven D Townsend
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, United States; Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, United States.
| |
Collapse
|
144
|
Hodzic Z, Bolock AM, Good M. The Role of Mucosal Immunity in the Pathogenesis of Necrotizing Enterocolitis. Front Pediatr 2017; 5:40. [PMID: 28316967 PMCID: PMC5334327 DOI: 10.3389/fped.2017.00040] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/15/2017] [Indexed: 12/29/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is the most devastating gastrointestinal disease of prematurity. Although the precise cause is not well understood, the main risk factors thought to contribute to NEC include prematurity, formula feeding, and bacterial colonization. Recent evidence suggests that NEC develops as a consequence of intestinal hyper-responsiveness to microbial ligands upon bacterial colonization in the preterm infant, initiating a cascade of aberrant signaling events, and a robust pro-inflammatory mucosal immune response. We now have a greater understanding of important mechanisms of disease pathogenesis, such as the role of cytokines, immunoglobulins, and immune cells in NEC. In this review, we will provide an overview of the mucosal immunity of the intestine and the relationship between components of the mucosal immune system involved in the pathogenesis of NEC, while highlighting recent advances in the field that have promise as potential therapeutic targets. First, we will describe the cellular components of the intestinal epithelium and mucosal immune system and their relationship to NEC. We will then discuss the relationship between the gut microbiota and cell signaling that underpins disease pathogenesis. We will conclude our discussion by highlighting notable therapeutic advancements in NEC that target the intestinal mucosal immunity.
Collapse
Affiliation(s)
- Zerina Hodzic
- University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Alexa M Bolock
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine , St. Louis, MO , USA
| | - Misty Good
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine , St. Louis, MO , USA
| |
Collapse
|
145
|
Oral supplementation of healthy adults with 2'-O-fucosyllactose and lacto-N-neotetraose is well tolerated and shifts the intestinal microbiota. Br J Nutr 2016; 116:1356-1368. [PMID: 27719686 PMCID: PMC5082288 DOI: 10.1017/s0007114516003354] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The gut microbiota has been established as an important player influencing many aspects
of human physiology. Breast milk, the first diet for an infant, contains human milk
oligosaccharides (HMO) that shape the infant’s gut microbiota by selectively stimulating
the growth of specific bacteria, especially bifidobacteria. In addition to their
bifidogenic activity, the ability of HMO to modulate immune function and the gut barrier
makes them prime candidates to restore a beneficial microbiota in dysbiotic adults and
provide health benefits. We conducted a parallel, double-blind, randomised,
placebo-controlled, HMO-supplementation study in 100 healthy, adult volunteers, consuming
chemically produced 2′-O-fucosyllactose (2′FL) and/or
lacto-N-neotetraose (LNnT) at various daily doses and mixes or placebo
for 2 weeks. All participants completed the study without premature discontinuation.
Supplementation of 2′FL and LNnT at daily doses up to 20 g was shown to be safe and well
tolerated, as assessed using the gastrointestinal symptoms rating scale. 16S rRNA
sequencing analysis showed that HMO supplementation specifically modified the adult gut
microbiota with the primary impact being substantial increases in relative abundance of
Actinobacteria and Bifidobacterium in particular and a reduction in
relative abundance of Firmicutes and Proteobacteria. This study provides the first set of
data on safety, tolerance and impact of HMO on the adult gut microbiota. Collectively, the
results from this study show that supplementing the diet with HMO is a valuable strategy
to shape the human gut microbiota and specifically promote the growth of beneficial
bifidobacteria.
Collapse
|
146
|
Biotechnological production of fucosylated human milk oligosaccharides: Prokaryotic fucosyltransferases and their use in biocatalytic cascades or whole cell conversion systems. J Biotechnol 2016; 235:61-83. [DOI: 10.1016/j.jbiotec.2016.03.052] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 03/30/2016] [Accepted: 03/31/2016] [Indexed: 01/29/2023]
|
147
|
Yu ZT, Nanthakumar NN, Newburg DS. The Human Milk Oligosaccharide 2'-Fucosyllactose Quenches Campylobacter jejuni-Induced Inflammation in Human Epithelial Cells HEp-2 and HT-29 and in Mouse Intestinal Mucosa. J Nutr 2016; 146:1980-1990. [PMID: 27629573 PMCID: PMC5037868 DOI: 10.3945/jn.116.230706] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 08/09/2016] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Campylobacter jejuni causes diarrhea worldwide; young children are most susceptible. Binding of virulent C. jejuni to the intestinal mucosa is inhibited ex vivo by α1,2-fucosylated carbohydrate moieties, including human milk oligosaccharides (HMOSs). OBJECTIVE The simplest α1,2-fucosylated HMOS structure, 2'-fucosyllactose (2'-FL), can be predominant at ≤5 g/L milk. Although 2'-FL inhibits C. jejuni binding ex vivo and in vivo, the effects of 2'FL on the cell invasion central to C. jejuni pathogenesis have not been tested. Clinical isolates of C. jejuni infect humans, birds, and ferrets, limiting studies on its mammalian pathobiology. METHODS Human epithelial cells HEp-2 and HT-29 infected with the virulent C. jejuni strain 81-176 human isolate were treated with 5 g 2'-FL/L, and the degree of infection and inflammatory response was measured. Four-week-old male wild-type C57BL/6 mice were fed antibiotics to reduce their intestinal microbiota and were inoculated with C. jejuni strain 81-176. The sensitivity of the resulting acute transient enteric infection and immune response to inhibition by 2'-FL ingestion was tested. RESULTS In HEp-2 and HT-29 cells, 2'-FL attenuated 80% of C. jejuni invasion (P < 0.05) and suppressed the release of mucosal proinflammatory signals of interleukin (IL) 8 by 60-70%, IL-1β by 80-90%, and the neutrophil chemoattractant macrophage inflammatory protein 2 (MIP-2) by 50% (P < 0.05). Ingestion of 2'-FL by mice reduced C. jejuni colonization by 80%, weight loss by 5%, histologic features of intestinal inflammation by 50-70%, and induction of inflammatory signaling molecules of the acute-phase mucosal immune response by 50-60% (P < 0.05). This acute model did not induce IL-17 (adaptive T cell response), a chronic response. CONCLUSIONS In human cells in vitro (HEp-2, HT-29) and in a mouse infection model that recapitulated key pathologic features of C. jejuni clinical disease, 2'-FL inhibited pathogenesis and its sequelae. These data strongly support the hypothesis that 2'-FL represents a new class of oral agent for prevention, and potentially for treatment, of specific enteric infectious diseases.
Collapse
Affiliation(s)
- Zhuo-Teng Yu
- Department of Biology, Boston College, Chestnut Hill, MA; and
| | - N Nanda Nanthakumar
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - David S Newburg
- Department of Biology, Boston College, Chestnut Hill, MA; and
| |
Collapse
|
148
|
Seppo AE, Autran CA, Bode L, Järvinen KM. Human milk oligosaccharides and development of cow's milk allergy in infants. J Allergy Clin Immunol 2016; 139:708-711.e5. [PMID: 27702672 DOI: 10.1016/j.jaci.2016.08.031] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/09/2016] [Accepted: 08/16/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Antti E Seppo
- Division of Allergy and Immunology and Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry and Golisano Children's Hospital, Rochester, NY.
| | | | - Lars Bode
- University of California San Diego, La Jolla, Calif
| | - Kirsi M Järvinen
- Division of Allergy and Immunology and Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry and Golisano Children's Hospital, Rochester, NY
| |
Collapse
|
149
|
Minimal short-term effect of dietary 2'-fucosyllactose on bacterial colonisation, intestinal function and necrotising enterocolitis in preterm pigs. Br J Nutr 2016; 116:834-41. [PMID: 27452119 DOI: 10.1017/s0007114516002646] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Human milk decreases the risk of necrotising enterocolitis (NEC), a severe gastrointestinal disease that occurs in 5-10 % of preterm infants. The prebiotic and immune-modulatory effects of milk oligosaccharides may contribute to this protection. Preterm pigs were used to test whether infant formula enriched with α1,2-fucosyllactose (2'-FL, the most abundant oligosaccharide in human milk) would benefit gut microbial colonisation and NEC resistance after preterm birth. Caesarean-delivered preterm pigs were fed formula (Controls, n 17) or formula with 5 g/l 2'-FL (2'-FL, n 16) for 5 d; eight 2'-FL pigs (50 %) and twelve Controls (71 %) developed NEC, with no difference in lesion scores (P=0·35); 2'-FL pigs tended to have less anaerobic bacteria in caecal contents (P=0·22), but no difference in gut microbiota between groups were observed by fluorescence in situ hybridisation and 454 pyrosequencing. Abundant α1,2-fucose was detected in the intestine with no difference between groups, and intestinal structure (villus height, permeability) and digestive function (hexose absorption, brush border enzyme activities) were not affected by 2'-FL. Formula enrichment with 2'-FL does not affect gut microbiology, digestive function or NEC sensitivity in pigs within the first few days after preterm birth. Milk 2'-FL may not be critical in the immediate postnatal period of preterm neonates when gut colonisation and intestinal immunity are still immature.
Collapse
|
150
|
Relative fermentation of oligosaccharides from human milk and plants by gut microbes. Eur Food Res Technol 2016. [DOI: 10.1007/s00217-016-2730-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|