1
|
Luo F, Zhang M, Zhang L, Zhou P. Nutritional and health effects of bovine colostrum in neonates. Nutr Rev 2024; 82:1631-1645. [PMID: 38052234 DOI: 10.1093/nutrit/nuad145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023] Open
Abstract
High concentrations of immunoglobulins, bioactive peptides, and growth factors are found in bovine colostrum (BC), the milk produced by cows in the first few days after parturition. Various biological functions make it increasingly used to provide nutritional support and immune protection to the offspring of many species, including humans. These biological functions include cell growth stimulation, anti-infection, and immunomodulation. The primary components and biological functions of colostrum were reviewed in the literature, and the authors also looked at its latent effects on the growth and development of neonates as well as on conditions such as infections, necrotizing enterocolitis, short bowel syndrome, and feeding intolerance. The importance of BC in neonatal nutrition, immune support, growth and development, and gut health has been demonstrated in a number of experimental and animal studies. BC has also been shown to be safe at low doses without adverse effects in newborns. BC supplementation has been shown to be efficient in preventing several disorders, including rotavirus diarrhea, necrotizing enterocolitis, and sepsis in animal models of prematurity and some newborn studies. Therefore, BC supplementation should be considered in cases where maternal milk is insufficient or donor milk is unavailable. The optimal age, timing, dosage, and form of BC administration still require further investigation.
Collapse
Affiliation(s)
- Fangmei Luo
- Department of Neonatology, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Min Zhang
- Department of Neonatology, Jinan University-Affiliated Shenzhen Baoan Women's and Children's Hospital, Shenzhen, China
| | - Lian Zhang
- Department of Neonatology, Jinan University-Affiliated Shenzhen Baoan Women's and Children's Hospital, Shenzhen, China
| | - Ping Zhou
- Department of Neonatology, Jinan University-Affiliated Shenzhen Baoan Women's and Children's Hospital, Shenzhen, China
| |
Collapse
|
2
|
Falsaperla R, Sortino V, Gambilonghi F, Vitaliti G, Striano P. Human Milk Oligosaccharides and Their Pivotal Role in Gut-Brain Axis Modulation and Neurologic Development: A Narrative Review to Decipher the Multifaceted Interplay. Nutrients 2024; 16:3009. [PMID: 39275324 PMCID: PMC11397282 DOI: 10.3390/nu16173009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 09/16/2024] Open
Abstract
BACKGROUND Human milk oligosaccharides (HMOs), which are unique bioactive components in human milk, are increasingly recognized for their multifaceted roles in infant health. A deeper understanding of the nexus between HMOs and the gut-brain axis can revolutionize neonatal nutrition and neurodevelopmental strategies. METHODS We performed a narrative review using PubMed, Embase, and Google Scholar to source relevant articles. The focus was on studies detailing the influence of HMOs on the gut and brain systems, especially in neonates. Articles were subsequently synthesized based on their exploration into the effects and mechanisms of HMOs on these interconnected systems. RESULTS HMOs significantly influence the neonatal gut-brain axis. Specific concentrations of HMO, measured 1 and 6 months after birth, would seem to agree with this hypothesis. HMOs are shown to influence gut microbiota composition and enhance neurotransmitter production, which are crucial for brain development. For instance, 2'-fucosyllactose has been demonstrated to support cognitive development by fostering beneficial gut bacteria that produce essential short-chain fatty acids. CONCLUSIONS HMOs serve as crucial modulators of the neonatal gut-brain axis, underscoring their importance in infant nutrition and neurodevelopment. Their dual role in shaping the infant gut while influencing brain function presents them as potential game-changers in neonatal health strategies.
Collapse
Affiliation(s)
- Raffaele Falsaperla
- Neonatal Intensive Care Unit and Neonatal Accompaniment Unit, Azienda Ospedaliero-Universitaria Policlinico "Rodolico-San Marco", San Marco Hospital, University of Catania, 95123 Catania, Italy
- Unit of Pediatrics and Pediatric Emergency, Azienda Ospedaliero-Universitaria Policlinico "Rodolico-San Marco", San Marco Hospital, University of Catania, 95123 Catania, Italy
- Department of Medical Science-Pediatrics, University of Ferrara, 44124 Ferrara, Italy
| | - Vincenzo Sortino
- Unit of Pediatrics and Pediatric Emergency, Azienda Ospedaliero-Universitaria Policlinico "Rodolico-San Marco", San Marco Hospital, University of Catania, 95123 Catania, Italy
| | - Francesco Gambilonghi
- Postgraduate Training Program in Pediatrics, Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
| | - Giovanna Vitaliti
- Unit of Pediatrics and Pediatric Emergency, Azienda Ospedaliero-Universitaria Policlinico "Rodolico-San Marco", San Marco Hospital, University of Catania, 95123 Catania, Italy
| | - Pasquale Striano
- IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy
| |
Collapse
|
3
|
Jepsen SD, Lund A, Matwiejuk M, Andresen L, Christensen KR, Skov S. Human milk oligosaccharides regulate human macrophage polarization and activation in response to Staphylococcus aureus. Front Immunol 2024; 15:1379042. [PMID: 38903508 PMCID: PMC11187579 DOI: 10.3389/fimmu.2024.1379042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/08/2024] [Indexed: 06/22/2024] Open
Abstract
Human milk oligosaccharides (HMOs) are present in high numbers in milk of lactating women. They are beneficial to gut health and the habitant microbiota, but less is known about their effect on cells from the immune system. In this study, we investigated the direct effect of three structurally different HMOs on human derived macrophages before challenge with Staphylococcus aureus (S. aureus). The study demonstrates that individual HMO structures potently affect the activation, differentiation and development of monocyte-derived macrophages in response to S. aureus. 6´-Sialyllactose (6'SL) had the most pronounced effect on the immune response against S. aureus, as illustrated by altered expression of macrophage surface markers, pointing towards an activated M1-like macrophage-phenotype. Similarly, 6'SL increased production of the pro-inflammatory cytokines TNF-α, IL-6, IL-8, IFN-γ and IL-1β, when exposing cells to 6'SL in combination with S. aureus compared with S. aureus alone. Interestingly, macrophages treated with 6'SL exhibited an altered proliferation profile and increased the production of the classic M1 transcription factor NF-κB. The HMOs also enhanced macrophage phagocytosis and uptake of S. aureus. Importantly, the different HMOs did not notably affect macrophage activation and differentiation without S. aureus exposure. Together, these findings show that HMOs can potently augment the immune response against S. aureus, without causing inflammatory activation in the absence of S. aureus, suggesting that HMOs assist the immune system in targeting important pathogens during early infancy.
Collapse
Affiliation(s)
- Stine Dam Jepsen
- dsm-firmenich, Hørsholm, Denmark
- Immunology, Section for Preclinical Disease Biology, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Astrid Lund
- Immunology, Section for Preclinical Disease Biology, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Lars Andresen
- Immunology, Section for Preclinical Disease Biology, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Søren Skov
- Immunology, Section for Preclinical Disease Biology, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
4
|
Boll EJ, Lopez DV, Terne M, Hessing S, Parschat K, Jensen SR. Human milk oligosaccharides differentially support gut barrier integrity and enhance Th1 and Th17 cell effector responses in vitro. Front Immunol 2024; 15:1359499. [PMID: 38510254 PMCID: PMC10950922 DOI: 10.3389/fimmu.2024.1359499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/15/2024] [Indexed: 03/22/2024] Open
Abstract
Human milk oligosaccharides (HMOs) can modulate the intestinal barrier and regulate immune cells to favor the maturation of the infant intestinal tract and immune system, but the precise functions of individual HMOs are unclear. To determine the structure-dependent effects of individual HMOs (representing different structural classes) on the intestinal epithelium as well as innate and adaptive immune cells, we assessed fucosylated (2'FL and 3FL), sialylated (3'SL and 6'SL) and neutral non-fucosylated (LNT and LNT2) HMOs for their ability to support intestinal barrier integrity, to stimulate the secretion of chemokines from intestinal epithelial cells, and to modulate cytokine release from LPS-activated dendritic cells (DCs), M1 macrophages (MØs), and co-cultures with naïve CD4+ T cells. The fucosylated and neutral non-fucosylated HMOs increased barrier integrity and protected the barrier following an inflammatory insult but exerted minimal immunomodulatory activity. The sialylated HMOs enhanced the secretion of CXCL10, CCL20 and CXCL8 from intestinal epithelial cells, promoted the secretion of several cytokines (including IL-10, IL-12p70 and IL-23) from LPS-activated DCs and M1 MØs, and increased the secretion of IFN-γ and IL-17A from CD4+ T cells primed by LPS-activated DCs and MØs while reducing the secretion of IL-13. Thus, 3'SL and 6'SL supported Th1 and Th17 responses while reducing Th2 responses. Collectively, our data show that HMOs exert structure-dependent effects on the intestinal epithelium and possess immunomodulatory properties that confer benefits to infants and possibly also later in life.
Collapse
Affiliation(s)
| | | | - Mandy Terne
- Chr. Hansen A/S, Applied HMOs, Hoersholm, Denmark
| | - Sara Hessing
- Chr. Hansen A/S, Applied HMOs, Hoersholm, Denmark
| | | | | |
Collapse
|
5
|
Monaco MH, Wang M, Hauser J, Yan J, Dilger RN, Donovan SM. Formula supplementation with human and bovine milk oligosaccharides modulates blood IgG and T-helper cell populations, and ex vivo LPS-stimulated cytokine production in a neonatal preclinical model. Front Immunol 2023; 14:1327853. [PMID: 38179055 PMCID: PMC10765566 DOI: 10.3389/fimmu.2023.1327853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/06/2023] [Indexed: 01/06/2024] Open
Abstract
Introduction Human milk contains structurally diverse oligosaccharides (HMO), which are multifunctional modulators of neonatal immune development. Our objective was to investigate formula supplemented with fucosylated (2'FL) + neutral (lacto-N-neotetraose, LNnt) oligosaccharides and/or sialylated bovine milk oligosaccharides (BMOS) on immunological outcomes. Methods Pigs (n=46) were randomized at 48h of age to four diets: sow milk replacer formula (CON), BMOS (CON + 6.5 g/L BMOS), HMO (CON + 1.0 g/L 2'FL + 0.5 g/L LNnT), or BMOS+HMO (CON + 6.5 g/L BMOS + 1.0 g/L 2'FL + 0.5 g/L LNnT). Blood and tissues were collected on postnatal day 33 for measurement of cytokines and IgG, phenotypic identification of immune cells, and ex vivo lipopolysaccharide (LPS)-stimulation of immune cells. Results Serum IgG was significantly lower in the HMO group than BMOS+HMO but did not differ from CON or BMOS. The percentage of PBMC T-helper cells was lower in BMOS+HMO than the other groups. Splenocytes from the BMOS group secreted more IL-1β when stimulated ex vivo with LPS compared to CON or HMO groups. For PBMCs, a statistical interaction of BMOS*HMO was observed for IL-10 secretion (p=0.037), with BMOS+HMO and HMO groups differing at p=0.1. Discussion The addition of a mix of fucosylated and sialylated oligosaccharides to infant formula provides specific activities in the immune system that differ from formulations supplemented with one oligosaccharide structure.
Collapse
Affiliation(s)
- Marcia H. Monaco
- Department of Food Science and Human Nutrition, University of Illinois at Urbana Champaign, Urbana, IL, United States
| | - Mei Wang
- Department of Food Science and Human Nutrition, University of Illinois at Urbana Champaign, Urbana, IL, United States
| | - Jonas Hauser
- Brain Health Department, Nestlé Institute of Health Sciences, Société des Produits Nestlé SA, Lausanne, Switzerland
| | - Jian Yan
- Nestlé Product Technology Center Nutrition, Vevey, Switzerland
| | - Ryan N. Dilger
- Department of Animal Sciences, University of Illinois at Urbana Champaign, Urbana, IL, United States
| | - Sharon M. Donovan
- Department of Food Science and Human Nutrition, University of Illinois at Urbana Champaign, Urbana, IL, United States
| |
Collapse
|
6
|
Tarrant I, Finlay BB. Human milk oligosaccharides: potential therapeutic aids for allergic diseases. Trends Immunol 2023:S1471-4906(23)00111-4. [PMID: 37438187 DOI: 10.1016/j.it.2023.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 07/14/2023]
Abstract
Childhood allergy, including asthma, eczema, and food allergies, is a major global health burden, with prevalence increasing dramatically and novel interventions needed. Emerging research suggests that human milk oligosaccharides (HMOs), complex glycans found in breastmilk, have allergy-protective properties, indicating exciting therapeutic potential. This review evaluates current literature on the role of HMOs in allergy, assesses underlying immunological mechanisms, and discusses future research needed to translate findings into clinical implications. HMOs may mediate allergy risk through multiple structure-specific mechanisms, including microbiome modification, intestinal barrier maturation, immunomodulation, and gene regulation. Findings emphasize the importance of breastfeeding encouragement and HMO-supplemented formula milk for high allergy-risk infants. Although further investigation is necessary to determine the most efficacious structures against varying allergy phenotypes and their long-term efficacy, HMOs may represent a promising complementary tool for childhood allergy prevention.
Collapse
Affiliation(s)
- Isabel Tarrant
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - B Brett Finlay
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada; Department of Biochemistry, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
7
|
Recent Research and Application Prospect of Functional Oligosaccharides on Intestinal Disease Treatment. Molecules 2022; 27:molecules27217622. [PMID: 36364447 PMCID: PMC9656564 DOI: 10.3390/molecules27217622] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/21/2022] [Accepted: 10/21/2022] [Indexed: 11/09/2022] Open
Abstract
The intestinal tract is an essential digestive organ of the human body, and damage to the intestinal barrier will lead to various diseases. Functional oligosaccharides are carbohydrates with a low degree of polymerization and exhibit beneficial effects on human intestinal health. Laboratory experiments and clinical studies indicate that functional oligosaccharides repair the damaged intestinal tract and maintain intestinal homeostasis by regulating intestinal barrier function, immune response, and intestinal microbial composition. Functional oligosaccharides treat intestinal disease such as inflammatory bowel disease (IBD) and colorectal cancer (CRC) and have excellent prospects for therapeutic application. Here, we present an overview of the recent research into the effects of functional oligosaccharides on intestinal health.
Collapse
|
8
|
Rosa F, Sharma AK, Gurung M, Casero D, Matazel K, Bode L, Simecka C, Elolimy AA, Tripp P, Randolph C, Hand TW, Williams KD, LeRoith T, Yeruva L. Human Milk Oligosaccharides Impact Cellular and Inflammatory Gene Expression and Immune Response. Front Immunol 2022; 13:907529. [PMID: 35844612 PMCID: PMC9278088 DOI: 10.3389/fimmu.2022.907529] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
Human milk harbors complex carbohydrates, including human milk oligosaccharides (HMOs), the third most abundant component after lactose and lipids. HMOs have been shown to impact intestinal microbiota, modulate the intestinal immune response, and prevent pathogenic bacterial binding by serving as decoy receptors. However, the direct effect of HMOs on intestinal function and immunity remains to be elucidated. To address this knowledge gap, 21-day-old germ-free mice (C57BI/6) were orally gavaged with 15 mg/day of pooled HMOs for 7 or 14 days and euthanized at day 28 or 35. A set of mice was maintained until day 50 to determine the persistent effects of HMOs. Control groups were maintained in the isolators for 28, 35, or 50 days of age. At the respective endpoints, intestinal tissues were subjected to histomorphometric and transcriptomic analyses, while the spleen and mesenteric lymph nodes (MLNs) were subjected to flow cytometric analysis. The small intestine (SI) crypt was reduced after HMO treatment relative to control at days 28 and 35, while the SI villus height and large intestine (LI) gland depth were decreased in the HMO-treated mice relative to the control at day 35. We report significant HMO-induced and location-specific gene expression changes in host intestinal tissues. HMO treatment significantly upregulated genes involved in extracellular matrix, protein ubiquitination, nuclear transport, and mononuclear cell differentiation. CD4+ T cells were increased in both MLNs and the spleen, while CD8+ T cells were increased in the spleen at day 50 in the HMO group in comparison to controls. In MLNs, plasma cells were increased in HMO group at days 28 and 35, while in the spleen, only at day 28 relative to controls. Macrophages/monocytes and neutrophils were lower in the spleen of the HMO group at days 28, 35, and 50, while in MLNs, only neutrophils were lower at day 50 in the 14-day HMO group. In addition, diphtheria toxoid and tetanus toxoid antibody-secreting cells were higher in HMO-supplemented group compared to controls. Our data suggest that HMOs have a direct effect on gastrointestinal tract metabolism and the immune system even in the absence of host microbiota.
Collapse
Affiliation(s)
- Fernanda Rosa
- Arkansas Children’s Nutrition Center, United States Department of Agriculture-Agricultural Research Service (USDA-ARS), Little Rock, AR, United States
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX, United States
| | - Ashok K. Sharma
- Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai, Los Angeles, CA, United States
| | - Manoj Gurung
- Arkansas Children’s Nutrition Center, United States Department of Agriculture-Agricultural Research Service (USDA-ARS), Little Rock, AR, United States
| | - David Casero
- Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai, Los Angeles, CA, United States
| | - Katelin Matazel
- Arkansas Children’s Nutrition Center, United States Department of Agriculture-Agricultural Research Service (USDA-ARS), Little Rock, AR, United States
| | - Lars Bode
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA, United States
- Department of Pediatrics, University of California San Diego, La Jolla, CA, United States
| | - Christy Simecka
- Division of Laboratory Animal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Ahmed A. Elolimy
- Arkansas Children’s Nutrition Center, United States Department of Agriculture-Agricultural Research Service (USDA-ARS), Little Rock, AR, United States
- Animal Production Department, National Research Centre, Giza, Egypt
| | - Patricia Tripp
- Arkansas Children’s Nutrition Center, United States Department of Agriculture-Agricultural Research Service (USDA-ARS), Little Rock, AR, United States
| | - Christopher Randolph
- Center for Translational Pediatric Research, Arkansas Children’s Research Institute, Little Rock, AR, United States
| | - Timothy W. Hand
- University of Pittsburgh School of Medicine, R.K. Mellon Foundation Institute for Pediatric Research, University of Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Keith D. Williams
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Tanya LeRoith
- Department of Biomedical Sciences & Pathobiology, Virginia Tech, Blacksburg, VA, United States
| | - Laxmi Yeruva
- Arkansas Children’s Nutrition Center, United States Department of Agriculture-Agricultural Research Service (USDA-ARS), Little Rock, AR, United States
| |
Collapse
|
9
|
Wang J, Chen MS, Wang RS, Hu JQ, Liu S, Wang YYF, Xing XL, Zhang BW, Liu JM, Wang S. Current Advances in Structure-Function Relationships and Dose-Dependent Effects of Human Milk Oligosaccharides. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:6328-6353. [PMID: 35593935 DOI: 10.1021/acs.jafc.2c01365] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
HMOs (human milk oligosaccharides) are the third most important nutrient in breast milk. As complex glycans, HMOs play an important role in regulating neonatal intestinal immunity, resisting viral and bacterial infections, displaying anti-inflammatory characteristics, and promoting brain development. Although there have been some previous reports of HMOs, a detailed literature review summarizing the structure-activity relationships and dose-dependent effects of HMOs is lacking. Hence, after introducing the structures and synthetic pathways of HMOs, this review summarizes and categorizes identified structure-function relationships of HMOs. Differential mechanisms of different structural HMOs utilization by microorganisms are summarized. This review also emphasizes the recent advances in the interactions between different health benefits and the variance of dosage effect based on in vitro cell tests, animal experiments, and human intervention studies. The potential relationships between the chemical structure, the dosage selection, and the physiological properties of HMOs as functional foods are vital for further understanding of HMOs and their future applications.
Collapse
Affiliation(s)
- Jin Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Meng-Shan Chen
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Rui-Shan Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Jia-Qiang Hu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Shuang Liu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Yuan-Yi-Fei Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Xiao-Long Xing
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Bo-Wei Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Jing-Min Liu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| |
Collapse
|
10
|
Interactions between human milk oligosaccharides, microbiota and immune factors in milk of women with and without mastitis. Sci Rep 2022; 12:1367. [PMID: 35079053 PMCID: PMC8789856 DOI: 10.1038/s41598-022-05250-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 12/20/2021] [Indexed: 12/21/2022] Open
Abstract
Lactational mastitis is an excellent target to study possible interactions between HMOs, immune factors and milk microbiota due to the infectious and inflammatory nature of this condition. In this work, microbiological, immunological and HMO profiles of milk samples from women with (MW) or without (HW) mastitis were compared. Secretor status in women (based on HMO profile) was not associated to mastitis. DFLNH, LNFP II and LSTb concentrations in milk were higher in samples from HW than from MW among Secretor women. Milk from HW was characterized by a low bacterial load (dominated by Staphylococcus epidermidis and streptococci), high prevalence of IL10 and IL13, and low sialylated HMO concentration. In contrast, high levels of staphylococci, streptococci, IFNγ and IL12 characterized milk from MW. A comparison between subacute (SAM) and acute (AM) mastitis cases revealed differences related to the etiological agent (S. epidermidis in SAM; Staphylococcus aureus in AM), milk immunological profile (high content of IL10 and IL13 in SAM and IL2 in AM) and milk HMOs profile (high content of 3FL in SAM and of LNT, LNnT, and LSTc in AM). These results suggest that microbiological, immunological and HMOs profiles of milk are related to mammary health of women.
Collapse
|
11
|
Abstract
The developing gut microbiome in infancy plays a key role in shaping the host immune system and metabolic state, and human milk is the main factor influencing its composition. Human milk does not only serve to feed the baby, but also to help the new-born adapt to its new environment and microbial exposures. Human milk protects the infant by providing multiple bioactive molecules, including human milk oligosaccharides (HMOs), which are the third most abundant solid component after lipids and lactose. The infant is unable to digest HMOs, so they reach the small and large intestines intact where they have many roles, including acting as prebiotics. Bifidobacterium spp. are the main, but not the only, commensals equipped with genes for HMO degradation. In this review we will outline the HMOs structures and functions, list the genes needed for their digestion, and describe the main strategies adopted by bacteria for their utilization.
Collapse
Affiliation(s)
- Andrea C Masi
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, 3rd Floor Leech Building, Newcastle NE2 4HH, UK
| | - Christopher J Stewart
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, 3rd Floor Leech Building, Newcastle NE2 4HH, UK
| |
Collapse
|
12
|
Wu RY, Li B, Horne RG, Ahmed A, Lee D, Robinson SC, Zhu H, Cadete M, Alganabi M, Filler R, Johnson-Henry KC, Delgado-Olguin P, Pierro A, Sherman PM. Structure-Function Relationships of Human Milk Oligosaccharides on the Intestinal Epithelial Transcriptome in Caco-2 Cells and a Murine Model of Necrotizing Enterocolitis. Mol Nutr Food Res 2021; 66:e2100893. [PMID: 34921749 DOI: 10.1002/mnfr.202100893] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/06/2021] [Indexed: 12/24/2022]
Abstract
SCOPE Necrotizing enterocolitis (NEC) is a devastating gastrointestinal emergency affecting preterm infants. Breastmilk protects against NEC, partly due to human milk oligosaccharides (HMOs). HMO compositions are highly diverse, and it is unclear if anti-NEC properties are specific to carbohydrate motifs. Here, this study compares intestinal epithelial transcriptomes of five synthetic HMOs (sHMOs) and examines structure-function relationships of HMOs on intestinal signaling. METHODS AND RESULTS This study interrogates the transcriptome of Caco-2Bbe1 cells in response to five synthetic HMOs (sHMOs) using RNA sequencing: 2'-fucosyllactose (2'-FL), 3-fucosyllactose (3FL), 6'-siallyllactose (6'-SL), lacto-N-tetraose (LNT), lacto-N-neotetraose (LNnT). Protection against intestinal barrier dysfunction and inflammation occurred in an HMO-dependent manner. Each sHMO exerts a unique set of host transcriptome changes and modulated unique signaling pathways. There is clustering between HMOs bearing similar side chains, with little overlap in gene regulation which is shared by all sHMOs. Interestingly, most sHMOs protect pups against NEC, exerting divergent mechanisms on intestinal cell morphology and inflammation. CONCLUSIONS These results demonstrate that while structurally distinct HMOs impact intestinal physiology, their mechanisms of action differ. This finding establishes the first structure-function relationship of HMOs in the context of intestinal cell signaling responses and offers a functional framework by which to screen and design HMO-like compounds.
Collapse
Affiliation(s)
- Richard Y Wu
- Cell Biology Program, The Hospital for Sick Children, Toronto, M5G 0A4, Canada.,Translational Medicine, Research Institute, Hospital for Sick Children, Toronto, M5G 0A4, Canada
| | - Bo Li
- Translational Medicine, Research Institute, Hospital for Sick Children, Toronto, M5G 0A4, Canada.,Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - Rachael G Horne
- Cell Biology Program, The Hospital for Sick Children, Toronto, M5G 0A4, Canada
| | - Abdalla Ahmed
- Translational Medicine, Research Institute, Hospital for Sick Children, Toronto, M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, M5S 1A8, Canada
| | - Dorothy Lee
- Translational Medicine, Research Institute, Hospital for Sick Children, Toronto, M5G 0A4, Canada
| | - Shaiya C Robinson
- Cell Biology Program, The Hospital for Sick Children, Toronto, M5G 0A4, Canada
| | - Haitao Zhu
- Translational Medicine, Research Institute, Hospital for Sick Children, Toronto, M5G 0A4, Canada.,Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, M5G 1X8, Canada.,Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Marissa Cadete
- Translational Medicine, Research Institute, Hospital for Sick Children, Toronto, M5G 0A4, Canada
| | - Mashriq Alganabi
- Translational Medicine, Research Institute, Hospital for Sick Children, Toronto, M5G 0A4, Canada
| | - Rachel Filler
- Translational Medicine, Research Institute, Hospital for Sick Children, Toronto, M5G 0A4, Canada
| | | | - Paul Delgado-Olguin
- Translational Medicine, Research Institute, Hospital for Sick Children, Toronto, M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, M5S 1A8, Canada.,Heart & Stroke Richard Lewar Center of Excellence, Toronto, M5S 3H2, Canada
| | - Agostino Pierro
- Translational Medicine, Research Institute, Hospital for Sick Children, Toronto, M5G 0A4, Canada.,Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, M5G 1X8, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, M5S 1A8, Canada
| | - Philip M Sherman
- Cell Biology Program, The Hospital for Sick Children, Toronto, M5G 0A4, Canada.,Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| |
Collapse
|
13
|
Rohsiswatmo R. Nutritional Management and Recommendation for Preterm Infants: A Narrative Review. AMERTA NUTRITION 2021. [DOI: 10.20473/amnt.v5i1sp.2021.1-13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background: Preterm birth is defined as birth before 37 completed weeks of pregnancy. It is the most important predictor of adverse health and development infant outcomes that extend into the early childhood and beyond. It is also the leading cause of childhood mortality under 5 years of age worldwide and responsible for approximately one million neonatal deaths. It is also a significant contributor to childhood morbidities, with many survivors are facing an increased risk of lifelong disability and poor quality of life. Purpose: In this article, we aimed to describe features of preterm infants, what makes them different from term infants, and what to consider in nutritional management of preterm infants through a traditional narrative literature review. Discussion: Preterm infants are predisposed to more health complications than term infants with higher morbidity and mortality. This morbidity and mortality can be reduced through timely interventions for the mother and the preterm infant. Maternal interventions, such as health education and administration of micronutrient supplementation, are given before or during pregnancy and at delivery, whereas appropriate care for the preterm infants should be initiated immediately after birth, which include early breastfeeding and optimalization of weight gain. Conclusion: Essential care of the preterm infants and early aggressive nutrition should be provided to support rapid growth that is associated with improved neurodevelopmental outcomes. The goal is not only about survival but making sure that these preterm infants grow and develop without any residual morbidity.
Collapse
|
14
|
Logtenberg MJ, Akkerman R, Hobé RG, Donners KMH, Van Leeuwen SS, Hermes GDA, de Haan BJ, Faas MM, Buwalda PL, Zoetendal EG, de Vos P, Schols HA. Structure-Specific Fermentation of Galacto-Oligosaccharides, Isomalto-Oligosaccharides and Isomalto/Malto-Polysaccharides by Infant Fecal Microbiota and Impact on Dendritic Cell Cytokine Responses. Mol Nutr Food Res 2021; 65:e2001077. [PMID: 34060703 PMCID: PMC8459273 DOI: 10.1002/mnfr.202001077] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/18/2021] [Indexed: 12/23/2022]
Abstract
SCOPE Next to galacto-oligosaccharides (GOS), starch-derived isomalto-oligosaccharide preparation (IMO) and isomalto/malto-polysaccharides (IMMP) could potentially be used as prebiotics in infant formulas. However, it remains largely unknown how the specific molecular structures of these non-digestible carbohydrates (NDCs) impact fermentability and immune responses in infants. METHODS AND RESULTS In vitro fermentation of GOS, IMO and IMMP using infant fecal inoculum of 2- and 8-week-old infants shows that only GOS and IMO are fermented by infant fecal microbiota. The degradation of GOS and IMO coincides with an increase in Bifidobacterium and production of acetate and lactate, which is more pronounced with GOS. Individual isomers with an (1↔1)-linkage or di-substituted reducing terminal glucose residue are more resistant to fermentation. GOS, IMO, and IMMP fermentation digesta attenuates cytokine profiles in immature dendritic cells (DCs), but the extent is dependent on the infants age and NDC structure. CONCLUSION The IMO preparation, containing reducing and non-reducing isomers, shows similar fermentation patterns as GOS in fecal microbiota of 2-week-old infants. Knowledge obtained on the substrate specificities of infant fecal microbiota and the subsequent regulatory effects of GOS, IMO and IMMP on DC responses might contribute to the design of tailored NDC mixtures for infants of different age groups.
Collapse
Affiliation(s)
- Madelon J. Logtenberg
- Laboratory of Food ChemistryWageningen University & ResearchBornse Weilanden 9Wageningen6708 WGThe Netherlands
| | - Renate Akkerman
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Centre GroningenGroningenThe Netherlands
| | - Rosan G. Hobé
- Laboratory of Food ChemistryWageningen University & ResearchBornse Weilanden 9Wageningen6708 WGThe Netherlands
| | - Kristel M. H. Donners
- Laboratory of Food ChemistryWageningen University & ResearchBornse Weilanden 9Wageningen6708 WGThe Netherlands
| | - Sander S. Van Leeuwen
- Cluster Human Nutrition & HealthDepartment of Laboratory MedicineUniversity Medical Center GroningenGroningenThe Netherlands
| | - Gerben D. A. Hermes
- Laboratory of MicrobiologyWageningen University & ResearchWageningenThe Netherlands
| | - Bart J. de Haan
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Centre GroningenGroningenThe Netherlands
| | - Marijke M. Faas
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Centre GroningenGroningenThe Netherlands
| | - Piet L. Buwalda
- Biobased Chemistry and TechnologyWageningen University & ResearchWageningenThe Netherlands
- Avebe Innovation CenterGroningenThe Netherlands
| | - Erwin G. Zoetendal
- Laboratory of MicrobiologyWageningen University & ResearchWageningenThe Netherlands
| | - Paul de Vos
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Centre GroningenGroningenThe Netherlands
| | - Henk A. Schols
- Laboratory of Food ChemistryWageningen University & ResearchBornse Weilanden 9Wageningen6708 WGThe Netherlands
| |
Collapse
|
15
|
Sangild PT, Vonderohe C, Melendez Hebib V, Burrin DG. Potential Benefits of Bovine Colostrum in Pediatric Nutrition and Health. Nutrients 2021; 13:nu13082551. [PMID: 34444709 PMCID: PMC8402036 DOI: 10.3390/nu13082551] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/02/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022] Open
Abstract
Bovine colostrum (BC), the first milk produced from cows after parturition, is increasingly used as a nutritional supplement to promote gut function and health in other species, including humans. The high levels of whey and casein proteins, immunoglobulins (Igs), and other milk bioactives in BC are adapted to meet the needs of newborn calves. However, BC supplementation may improve health outcomes across other species, especially when immune and gut functions are immature in early life. We provide a review of BC composition and its effects in infants and children in health and selected diseases (diarrhea, infection, growth-failure, preterm birth, necrotizing enterocolitis (NEC), short-bowel syndrome, and mucositis). Human trials and animal studies (mainly in piglets) are reviewed to assess the scientific evidence of whether BC is a safe and effective antimicrobial and immunomodulatory nutritional supplement that reduces clinical complications related to preterm birth, infections, and gut disorders. Studies in infants and animals suggest that BC should be supplemented at an optimal age, time, and level to be both safe and effective. Exclusive BC feeding is not recommended for infants because of nutritional imbalances relative to human milk. On the other hand, adverse effects, including allergies and intolerance, appear unlikely when BC is provided as a supplement within normal nutrition guidelines for infants and children. Larger clinical trials in infant populations are needed to provide more evidence of health benefits when patients are supplemented with BC in addition to human milk or formula. Igs and other bioactive factors in BC may work in synergy, making it critical to preserve bioactivity with gentle processing and pasteurization methods. BC has the potential to become a safe and effective nutritional supplement for several pediatric subpopulations.
Collapse
Affiliation(s)
- Per Torp Sangild
- Comparative Pediatrics & Nutrition, University of Copenhagen, DK-1870 Copenhagen, Denmark;
- Department of Neonatology, Rigshospitalet, DK-1870 Copenhagen, Denmark
- Department of Pediatrics, Odense University Hospital, DK-5000 Odense, Denmark
| | - Caitlin Vonderohe
- USDA-ARS Children’s Nutrition Research Center, Pediatrics, Gastroenterology & Nutrition, Baylor College of Medicine, Houston, TX 77030, USA; (C.V.); (V.M.H.)
| | - Valeria Melendez Hebib
- USDA-ARS Children’s Nutrition Research Center, Pediatrics, Gastroenterology & Nutrition, Baylor College of Medicine, Houston, TX 77030, USA; (C.V.); (V.M.H.)
| | - Douglas G. Burrin
- USDA-ARS Children’s Nutrition Research Center, Pediatrics, Gastroenterology & Nutrition, Baylor College of Medicine, Houston, TX 77030, USA; (C.V.); (V.M.H.)
- Correspondence: ; Tel.: +1-713-798-7049
| |
Collapse
|
16
|
Rousseaux A, Brosseau C, Le Gall S, Piloquet H, Barbarot S, Bodinier M. Human Milk Oligosaccharides: Their Effects on the Host and Their Potential as Therapeutic Agents. Front Immunol 2021; 12:680911. [PMID: 34108974 PMCID: PMC8180913 DOI: 10.3389/fimmu.2021.680911] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022] Open
Abstract
Breastmilk is known to be very important for infants because it provides nutrients and immunological compounds. Among these compounds, human milk oligosaccharides (HMOs) represent the third most important component of breastmilk after lipids and lactose. Several experiments demonstrated the beneficial effects of these components on the microbiota, the immune system and epithelial barriers, which are three major biological systems. Indeed, HMOs induce bacterial colonization in the intestinal tract, which is beneficial for health. The gut bacteria can act directly and indirectly on the immune system by stimulating innate immunity and controlling inflammatory reactions and by inducing an adaptive immune response and a tolerogenic environment. In parallel, HMOs directly strengthen the intestinal epithelial barrier, protecting the host against pathogens. Here, we review the molecular mechanisms of HMOs in these different compartments and highlight their potential use as new therapeutic agents, especially in allergy prevention.
Collapse
Affiliation(s)
- Anaïs Rousseaux
- INRAE, Biopolyméres Interactions Assemblages, Nantes, France
| | - Carole Brosseau
- INRAE, Biopolyméres Interactions Assemblages, Nantes, France
| | - Sophie Le Gall
- INRAE, Biopolyméres Interactions Assemblages, Nantes, France.,INRAE, Bioressources: Imagerie, Biochimie & Structure, Nantes, France
| | - Hugues Piloquet
- Centre Hospitalier Universitaire Nantes, UMR1280 PhAN, Nantes, France
| | | | - Marie Bodinier
- INRAE, Biopolyméres Interactions Assemblages, Nantes, France
| |
Collapse
|
17
|
Bovine Milk Oligosaccharides and Human Milk Oligosaccharides Modulate the Gut Microbiota Composition and Volatile Fatty Acid Concentrations in a Preclinical Neonatal Model. Microorganisms 2021; 9:microorganisms9050884. [PMID: 33919138 PMCID: PMC8143120 DOI: 10.3390/microorganisms9050884] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
Milk oligosaccharides (OS) shape microbiome structure and function, but their relative abundances differ between species. Herein, the impact of the human milk oligosaccharides (HMO) (2′-fucosyllactose [2′FL] and lacto-N-neotetraose [LNnT]) and OS isolated from bovine milk (BMOS) on microbiota composition and volatile fatty acid (VFA) concentrations in ascending colon (AC) contents and feces was assessed. Intact male piglets received diets either containing 6.5 g/L BMOS (n = 12), 1.0 g/L 2′FL + 0.5 g/L LNnT (HMO; n = 12), both (HMO + BMOS; n = 10), or neither (CON; n = 10) from postnatal day (PND) 2 to 34. Microbiota were assessed by 16S rRNA gene sequencing and real-time PCR, and VFA were measured by gas chromatography. The microbiota was affected by OS in an intestine region-specific manner. BMOS reduced (p < 0.05) microbial richness in the AC, microbiota composition in the AC and feces, and acetate concentrations in AC, regardless of HMO presence. HMO alone did not affect overall microbial composition, but increased (p < 0.05) the relative proportion of specific taxa, including Blautia, compared to other groups. Bacteroides abundance was increased (p < 0.05) in the AC by BMOS and synergistically by BMOS + HMO in the feces. Distinct effects of HMO and BMOS suggest complementary and sometimes synergistic benefits of supplementing a complex mixture of OS to formula.
Collapse
|
18
|
Moubareck CA. Human Milk Microbiota and Oligosaccharides: A Glimpse into Benefits, Diversity, and Correlations. Nutrients 2021; 13:1123. [PMID: 33805503 PMCID: PMC8067037 DOI: 10.3390/nu13041123] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/18/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
Human milk represents a cornerstone for growth and development of infants, with extensive array of benefits. In addition to exceptionally nutritive and bioactive components, human milk encompasses a complex community of signature bacteria that helps establish infant gut microbiota, contributes to maturation of infant immune system, and competitively interferes with pathogens. Among bioactive constituents of milk, human milk oligosaccharides (HMOs) are particularly significant. These are non-digestible carbohydrates forming the third largest solid component in human milk. Valuable effects of HMOs include shaping intestinal microbiota, imparting antimicrobial effects, developing intestinal barrier, and modulating immune response. Moreover, recent investigations suggest correlations between HMOs and milk microbiota, with complex links possibly existing with environmental factors, genetics, geographical location, and other factors. In this review, and from a physiological and health implications perspective, milk benefits for newborns and mothers are highlighted. From a microbiological perspective, a focused insight into milk microbiota, including origins, diversity, benefits, and effect of maternal diet is presented. From a metabolic perspective, biochemical, physiological, and genetic significance of HMOs, and their probable relations to milk microbiota, are addressed. Ongoing research into mechanistic processes through which the rich biological assets of milk promote development, shaping of microbiota, and immunity is tackled.
Collapse
Affiliation(s)
- Carole Ayoub Moubareck
- College of Natural and Health Sciences, Zayed University, Dubai 19282, United Arab Emirates
| |
Collapse
|
19
|
In Love with Shaping You-Influential Factors on the Breast Milk Content of Human Milk Oligosaccharides and Their Decisive Roles for Neonatal Development. Nutrients 2020; 12:nu12113568. [PMID: 33233832 PMCID: PMC7699834 DOI: 10.3390/nu12113568] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/08/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023] Open
Abstract
Human milk oligosaccharides (HMOs) are structurally versatile sugar molecules constituting the third major group of soluble components in human breast milk. Based on the disaccharide lactose, the mammary glands of future and lactating mothers produce a few hundreds of different HMOs implicating that their overall anabolism utilizes rather high amounts of energy. At first sight, it therefore seems contradictory that these sugars are indigestible for infants raising the question of why such an energy-intensive molecular class evolved. However, in-depth analysis of their molecular modes of action reveals that Mother Nature created HMOs for neonatal development, protection and promotion of health. This is not solely facilitated by HMOs in their indigestible form but also by catabolites that are generated by microbial metabolism in the neonatal gut additionally qualifying HMOs as natural prebiotics. This narrative review elucidates factors influencing the HMO composition as well as physiological roles of HMOs on their way through the infant body and within the gut, where a major portion of HMOs faces microbial catabolism. Concurrently, this work summarizes in vitro, preclinical and observational as well as interventional clinical studies that analyzed potential health effects that have been demonstrated by or were related to either human milk-derived or synthetic HMOs or HMO fractions.
Collapse
|
20
|
Al-Khafaji AH, Jepsen SD, Christensen KR, Vigsnæs LK. The potential of human milk oligosaccharides to impact the microbiota-gut-brain axis through modulation of the gut microbiota. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104176] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
21
|
Šuligoj T, Vigsnæs LK, den Abbeele PV, Apostolou A, Karalis K, Savva GM, McConnell B, Juge N. Effects of Human Milk Oligosaccharides on the Adult Gut Microbiota and Barrier Function. Nutrients 2020; 12:E2808. [PMID: 32933181 PMCID: PMC7551690 DOI: 10.3390/nu12092808] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022] Open
Abstract
Human milk oligosaccharides (HMOs) shape the gut microbiota in infants by selectively stimulating the growth of bifidobacteria. Here, we investigated the impact of HMOs on adult gut microbiota and gut barrier function using the Simulator of the Human Intestinal Microbial Ecosystem (SHIME®), Caco2 cell lines, and human intestinal gut organoid-on-chips. We showed that fermentation of 2'-O-fucosyllactose (2'FL), lacto-N-neotetraose (LNnT), and combinations thereof (MIX) led to an increase of bifidobacteria, accompanied by an increase of short chain fatty acid (SCFA), in particular butyrate with 2'FL. A significant reduction in paracellular permeability of FITC-dextran probe was observed using Caco2 cell monolayers with fermented 2'FL and MIX, which was accompanied by an increase in claudin-8 gene expression as shown by qPCR, and a reduction in IL-6 as determined by multiplex ELISA. Using gut-on-chips generated from human organoids derived from proximal, transverse, and distal colon biopsies (Colon Intestine Chips), we showed that claudin-5 was significantly upregulated across all three gut-on-chips following treatment with fermented 2'FL under microfluidic conditions. Taken together, these data show that, in addition to their bifidogenic activity, HMOs have the capacity to modulate immune function and the gut barrier, supporting the potential of HMOs to provide health benefits in adults.
Collapse
Affiliation(s)
- Tanja Šuligoj
- Quadram Institute Bioscience, Gut Microbes and Health Institute Strategic Programme, Norwich Research Park, Norwich NR4 7UQ, UK;
| | | | | | - Athanasia Apostolou
- Emulate Inc., 27 Drydock Ave, Boston, MA 02210, USA; (A.A.); (K.K.)
- Graduate Program, Department of Medicine, School of Health Sciences, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Katia Karalis
- Emulate Inc., 27 Drydock Ave, Boston, MA 02210, USA; (A.A.); (K.K.)
| | - George M. Savva
- Quadram Institute Bioscience, Core Sciences Resources, Norwich Research Park, Norwich NR5 7UQ, UK;
| | - Bruce McConnell
- Glycom A/S, Kogle Allé 4, DK-2970 Hørsholm, Denmark; (L.K.V.); (B.M.)
| | - Nathalie Juge
- Quadram Institute Bioscience, Gut Microbes and Health Institute Strategic Programme, Norwich Research Park, Norwich NR4 7UQ, UK;
| |
Collapse
|
22
|
Bondue P, Milani C, Arnould E, Ventura M, Daube G, LaPointe G, Delcenserie V. Bifidobacterium mongoliense genome seems particularly adapted to milk oligosaccharide digestion leading to production of antivirulent metabolites. BMC Microbiol 2020; 20:111. [PMID: 32380943 PMCID: PMC7206731 DOI: 10.1186/s12866-020-01804-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 04/26/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Human milk oligosaccharides (HMO) could promote the growth of bifidobacteria, improving young children's health. In addition, fermentation of carbohydrates by bifidobacteria can result in the production of metabolites presenting an antivirulent activity against intestinal pathogens. Bovine milk oligosaccharides (BMO), structurally similar to HMO, are found at high concentration in cow whey. This is particularly observed for 3'-sialyllactose (3'SL). This study focused on enzymes and transport systems involved in HMO/BMO metabolism contained in B. crudilactis and B. mongoliense genomes, two species from bovine milk origin. The ability of B. mongoliense to grow in media supplemented with whey or 3'SL was assessed. Next, the effects of cell-free spent media (CFSM) were tested against the virulence expression of Escherichia coli O157:H7 and Salmonella enterica serovar Typhimurium. RESULTS Due to the presence of genes encoding β-galactosidases, β-hexosaminidases, α-sialidases and α-fucosidases, B. mongoliense presents a genome more sophisticated and more adapted to the digestion of BMO/HMO than B. crudilactis (which contains only β-galactosidases). In addition, HMO/BMO digestion involves genes encoding oligosaccharide transport systems found in B. mongoliense but not in B. crudilactis. B. mongoliense seemed able to grow on media supplemented with whey or 3'SL as main source of carbon (8.3 ± 1.0 and 6.7 ± 0.3 log cfu/mL, respectively). CFSM obtained from whey resulted in a significant under-expression of ler, fliC, luxS, stx1 and qseA genes (- 2.2, - 5.3, - 2.4, - 2.5 and - 4.8, respectively; P < 0.05) of E. coli O157:H7. CFSM from 3'SL resulted in a significant up-regulation of luxS (2.0; P < 0.05) gene and a down-regulation of fliC (- 5.0; P < 0.05) gene. CFSM obtained from whey resulted in significant up-regulations of sopD and hil genes (2.9 and 3.5, respectively; P < 0.05) of S. Typhimurium, while CFSM obtained from 3'SL fermentation down-regulated hil and sopD genes (- 2.7 and - 4.2, respectively; P < 0.05). CONCLUSION From enzymes and transporters highlighted in the genome of B. mongoliense and its potential ability to metabolise 3'SL and whey, B. mongoliense seems well able to digest HMO/BMO. The exact nature of the metabolites contained in CFSM has to be identified still. These results suggest that BMO associated with B. mongoliense could be an interesting synbiotic formulation to maintain or restore intestinal health of young children.
Collapse
Affiliation(s)
- Pauline Bondue
- Department of Food Science, Fundamental and Applied Research for Animal and Health, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Emilie Arnould
- Department of Food Science, Fundamental and Applied Research for Animal and Health, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Georges Daube
- Department of Food Science, Fundamental and Applied Research for Animal and Health, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Gisèle LaPointe
- Canadian Research Institute for Food Safety, University of Guelph, Guelph, Canada
| | - Véronique Delcenserie
- Department of Food Science, Fundamental and Applied Research for Animal and Health, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium.
| |
Collapse
|
23
|
Ali F, Lui K, Wang A, Day AS, Leach ST. The perinatal period, the developing intestinal microbiome and inflammatory bowel diseases: What links early life events with later life disease? J R Soc N Z 2020. [DOI: 10.1080/03036758.2019.1706586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Fathalla Ali
- School of Women’s and Children’s Health, University of New South Wales, Sydney, Australia
| | - Kei Lui
- School of Women’s and Children’s Health, University of New South Wales, Sydney, Australia
- Department of Newborn Care, Royal Hospital for Women, Sydney, Australia
| | - Alex Wang
- Faculty of Health, University of Technology Sydney, Sydney, Australia
| | - Andrew S. Day
- Department of Paediatrics, University of Otago, Christchurch, New Zealand
| | - Steven T. Leach
- School of Women’s and Children’s Health, University of New South Wales, Sydney, Australia
| |
Collapse
|
24
|
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: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
25
|
Human milk oligosaccharides and its acid hydrolysate LNT2 show immunomodulatory effects via TLRs in a dose and structure-dependent way. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.05.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
26
|
Morrin ST, Owens RA, Le Berre M, Gerlach JQ, Joshi L, Bode L, Irwin JA, Hickey RM. Interrogation of Milk-Driven Changes to the Proteome of Intestinal Epithelial Cells by Integrated Proteomics and Glycomics. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:1902-1917. [PMID: 30663306 DOI: 10.1021/acs.jafc.8b06484] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Bovine colostrum is a rich source of bioactive components which are important in the development of the intestine, in stimulating gut structure and function and in preparing the gut surface for subsequent colonization of microbes. What is not clear, however, is how colostrum may affect the repertoire of receptors and membrane proteins of the intestinal surface and the post-translational modifications associated with them. In the present work, we aimed to characterize the surface receptor and glycan profile of human HT-29 intestinal cells after exposure to a bovine colostrum fraction (BCF) by means of proteomic and glycomic analyses. Integration of label-free quantitative proteomic analysis and lectin array profiles confirmed that BCF exposure results in changes in the levels of glycoproteins present at the cell surface and also changes to their glycosylation pattern. This study contributes to our understanding of how milk components may regulate intestinal cells and prime them for bacterial interaction.
Collapse
Affiliation(s)
- Sinead T Morrin
- Teagasc Food Research Centre , Moorepark , Fermoy, P61C996 , County Cork , Ireland
- Veterinary Sciences Centre, School of Veterinary Medicine , University College Dublin , Belfield, Dublin 4, D04 V1W8 , Ireland
| | - Rebecca A Owens
- Department of Biology , Maynooth University , Maynooth , W23 F2H6 , County Kildare , Ireland
| | - Marie Le Berre
- Glycoscience Group, Advanced Glycoscience Research Cluster, National Centre for Biomedical Engineering Science , National University of Ireland Galway , H91TK33 , Galway , Ireland
| | - Jared Q Gerlach
- Glycoscience Group, Advanced Glycoscience Research Cluster, National Centre for Biomedical Engineering Science , National University of Ireland Galway , H91TK33 , Galway , Ireland
| | - Lokesh Joshi
- Glycoscience Group, Advanced Glycoscience Research Cluster, National Centre for Biomedical Engineering Science , National University of Ireland Galway , H91TK33 , Galway , Ireland
| | - Lars Bode
- Department of Pediatrics and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence , University of California, San Diego , La Jolla , California 92093 , United States
| | - Jane A Irwin
- Veterinary Sciences Centre, School of Veterinary Medicine , University College Dublin , Belfield, Dublin 4, D04 V1W8 , Ireland
| | - Rita M Hickey
- Teagasc Food Research Centre , Moorepark , Fermoy, P61C996 , County Cork , Ireland
| |
Collapse
|
27
|
Sialylated Oligosaccharides and Glycoconjugates of Human Milk. The Impact on Infant and Newborn Protection, Development and Well-Being. Nutrients 2019; 11:nu11020306. [PMID: 30717166 PMCID: PMC6413137 DOI: 10.3390/nu11020306] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 01/25/2019] [Accepted: 01/29/2019] [Indexed: 01/19/2023] Open
Abstract
Human milk not only has nutritional value, but also provides a wide range of biologically active molecules, which are adapted to meet the needs of newborns and infants. Mother’s milk is a source of sialylated oligosaccharides and glycans that are attached to proteins and lipids, whose concentrations and composition are unique. Sialylated human milk glycoconjugates and oligosaccharides enrich the newborn immature immune system and are crucial for their proper development and well-being. Some of the milk sialylated oligosaccharide structures can locally exert biologically active effects in the newborn’s and infant’s gut. Sialylated molecules of human milk can be recognized and bound by sialic acid-dependent pathogens and inhibit their adhesion to the epithelial cells of newborns and infants. A small amount of intact sialylated oligosaccharides can be absorbed from the intestine and remain in the newborn’s circulation in concentrations high enough to modulate the immunological system at the cellular level and facilitate proper brain development during infancy. Conclusion: The review summarizes the current state of knowledge on sialylated human milk oligosaccharides and glycoconjugates, discusses the significance of sialylated structures of human milk in newborn protection and development, and presents the advantages of human milk over infant formula.
Collapse
|
28
|
Bovine colostrum-driven modulation of intestinal epithelial cells for increased commensal colonisation. Appl Microbiol Biotechnol 2019; 103:2745-2758. [PMID: 30685814 DOI: 10.1007/s00253-019-09642-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/04/2019] [Accepted: 01/10/2019] [Indexed: 01/04/2023]
Abstract
Nutritional intake may influence the intestinal epithelial glycome and in turn the available attachment sites for bacteria. In this study, we tested the hypothesis that bovine colostrum may influence the intestinal cell surface and in turn the attachment of commensal organisms. Human HT-29 intestinal cells were exposed to a bovine colostrum fraction (BCF) rich in free oligosaccharides. The adherence of several commensal bacteria, comprising mainly bifidobacteria, to the intestinal cells was significantly enhanced (up to 52-fold) for all strains tested which spanned species that are found across the human lifespan. Importantly, the changes to the HT-29 cell surface did not support enhanced adhesion of the enteric pathogens tested. The gene expression profile of the HT-29 cells following treatment with the BCF was evaluated by microarray analysis. Many so called "glyco-genes" (glycosyltransferases and genes involved in the complex biosynthetic pathways of glycans) were found to be differentially regulated suggesting modulation of the enzymatic addition of sugars to glycoconjugate proteins. The microarray data was further validated by means of real-time PCR. The current findings provide an insight into how commensal microorganisms colonise the human gut and highlight the potential of colostrum and milk components as functional ingredients that can potentially increase commensal numbers in individuals with lower counts of health-promoting bacteria.
Collapse
|
29
|
Plaza-Díaz J, Fontana L, Gil A. Human Milk Oligosaccharides and Immune System Development. Nutrients 2018; 10:1038. [PMID: 30096792 PMCID: PMC6116142 DOI: 10.3390/nu10081038] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 08/01/2018] [Accepted: 08/06/2018] [Indexed: 12/26/2022] Open
Abstract
Maternal milk contains compounds that may affect newborn immunity. Among these are a group of oligosaccharides that are synthesized in the mammary gland from lactose; these oligosaccharides have been termed human milk oligosaccharides (HMOs). The amount of HMOs present in human milk is greater than the amount of protein. In fact, HMOs are the third-most abundant solid component in maternal milk after lactose and lipids, and are thus considered to be key components. The importance of HMOs may be explained by their inhibitory effects on the adhesion of microorganisms to the intestinal mucosa, the growth of pathogens through the production of bacteriocins and organic acids, and the expression of genes that are involved in inflammation. This review begins with short descriptions of the basic structures of HMOs and the gut immune system, continues with the beneficial effects of HMOs shown in cell and animal studies, and it ends with the observational and randomized controlled trials carried out in humans to date, with particular emphasis on their effect on immune system development. HMOs seem to protect breastfed infants against microbial infections. The protective effect has been found to be exerted through cell signaling and cell-to-cell recognition events, enrichment of the protective gut microbiota, the modulation of microbial adhesion, and the invasion of the infant intestinal mucosa. In addition, infants fed formula supplemented with selected HMOs exhibit a pattern of inflammatory cytokines closer to that of exclusively breastfed infants. Unfortunately, the positive effects found in preclinical studies have not been substantiated in the few randomized, double-blinded, multicenter, controlled trials that are available, perhaps partly because these studies focus on aspects other than the immune response (e.g., growth, tolerance, and stool microbiota).
Collapse
Affiliation(s)
- Julio Plaza-Díaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain.
- Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center, Parque Tecnológico Ciencias de la Salud, University of Granada, Armilla, 18100 Granada, Spain.
- Instituto de Investigación Biosanitaria ibs., 18014 Granada, Spain.
| | - Luis Fontana
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain.
- Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center, Parque Tecnológico Ciencias de la Salud, University of Granada, Armilla, 18100 Granada, Spain.
- Instituto de Investigación Biosanitaria ibs., 18014 Granada, Spain.
| | - Angel Gil
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain.
- Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center, Parque Tecnológico Ciencias de la Salud, University of Granada, Armilla, 18100 Granada, Spain.
- Instituto de Investigación Biosanitaria ibs., 18014 Granada, Spain.
- CIBEROBN, Instituto de Salud Carlos III, 28029 Madrid, Spain.
| |
Collapse
|
30
|
Dai Z, Feng S, Liu A, Wang H, Zeng X, Yang CS. Anti-inflammatory effects of newly synthesized α-galacto-oligosaccharides on dextran sulfate sodium-induced colitis in C57BL/6J mice. Food Res Int 2018; 109:350-357. [PMID: 29803460 PMCID: PMC5976246 DOI: 10.1016/j.foodres.2018.04.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/12/2018] [Accepted: 04/23/2018] [Indexed: 12/27/2022]
Abstract
α-Galacto-oligosaccharides have been reported to have beneficial health effects. The purpose of this study was to investigate the preventive effects of a newly synthesized α-galacto-oligosaccharide mixture (α-GOSg), as well as raffinose family oligosaccharides (RFOs), on dextran sulfate sodium (DSS)-induced colitis in mice. When administered in drinking water at 0.5% for 15 days, both α-GOSg and RFOs significantly decreased fecal hemoglobin content, partially prevented colon length shortening, reduced the severity of colon inflammation, and attenuated DSS-induced upregulation of cyclooxygenase-2. In addition, the activation of the inflammatory regulator nuclear factor-kappa B was slightly inhibited by α-GOSg. The results showed that the newly synthesized α-GOSg preparation has similar anti-inflammatory activities as RFOs in this colitis model. The anti-inflammatory activity of α-GOSg in humans remains to be investigated.
Collapse
Affiliation(s)
- Zhuqing Dai
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Simin Feng
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Anna Liu
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Hong Wang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Xiaoxiong Zeng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Chung S Yang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.
| |
Collapse
|
31
|
O'Sullivan A, Salcedo J, Rubert J. Advanced analytical strategies for measuring free bioactive milk sugars: from composition and concentrations to human metabolic response. Anal Bioanal Chem 2018. [PMID: 29536151 DOI: 10.1007/s00216-018-0913-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Our daily food intake provides the nutrients to maintain health. However, in addition to the nutritional values, food can promote health and be beneficial in preventing diseases. Human milk is a unique food source that contains essential nutrients in the right balance and other bioactive factors that make it the ideal food for all healthy term infants. Human milk oligosaccharides (HMOs) play an important role in health, at several levels: acting as prebiotics promoting the growth of beneficial bacterial strains, preventing the growth of pathogenic bacteria in the intestine, and modulating the immune response against bacterial infections. However, despite their biological relevance and the advances made in the analytical field, very few studies have been carried out to better understand HMOs bioactivity mechanisms or to examine human metabolic response to dietary supplementation. This review describes the state-of-the-art of glycomics strategies, recent analytical methods, and future trends for the identification and discovery of bioactive sugars, the known mechanisms of action, and discusses findings of some recent human intervention trials.
Collapse
Affiliation(s)
- Aifric O'Sullivan
- UCD Institute for Food and Health, University College Dublin, Belfield, Dublin 4, Ireland
| | - Jaime Salcedo
- Department of Food Science & Technology, University of California-Davis, One Shields Av, Davis, CA, 95616, USA.,Chemistry Product Development, Waters Technologies Ireland Ltd., Wexford Business Park, Drinagh, Ireland
| | - Josep Rubert
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach (FEM), Via E. Mach 1, 38010, San Michele all'Adige, Italy.
| |
Collapse
|
32
|
Morozov V, Hansman G, Hanisch FG, Schroten H, Kunz C. Human Milk Oligosaccharides as Promising Antivirals. Mol Nutr Food Res 2018; 62:e1700679. [PMID: 29336526 DOI: 10.1002/mnfr.201700679] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/23/2017] [Indexed: 11/07/2022]
Abstract
Human milk oligosaccharides (HMOs) are diverse unconjugated carbohydrates that are highly abundant in human breast milk. These glycans are investigated in the context of exhibiting multiple functions in infant growth and development. They seem to provide protection against infectious diseases, including a number of poorly manageable viral infections. Although the potential mechanism of the HMO antiviral protection is rather broad, much of the current experimental work has focused on studying of HMO antiadhesive properties. HMOs may mimic structures of viral receptors and block adherence to target cells, thus preventing infection. Still, the potential of HMOs as a source for new antiviral drugs is relatively unexploited. This can be partly attributed to the extreme complexity of the virus-carbohydrate interactions and technical difficulties in HMO isolation, characterization, and manufacturing procedures. Fortunately, we are currently entering a period of major technological advances that have enabled deeper insights into carbohydrate mediated viral entry, rational selection of HMOs as anti-entry inhibitors, and even evaluation of individual synthetic HMO structures. Here, we provide an up-to-date review on glycan binding studies for rotaviruses, noroviruses, influenza viruses, and human immunodeficiency viruses. We also discuss the preventive and therapeutic potential of HMOs as anti-entry inhibitors and address challenges on the route from fundamental studies to clinical trials.
Collapse
Affiliation(s)
- Vasily Morozov
- Pediatric Infectious Diseases Unit, University Children's Hospital Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Grant Hansman
- Schaller Research Group at the University of Heidelberg and the DKFZ, Heidelberg, Germany
| | - Franz-Georg Hanisch
- Institute of Biochemistry II, Medical Faculty, University of Cologne, Cologne, Germany
| | - Horst Schroten
- Pediatric Infectious Diseases Unit, University Children's Hospital Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Clemens Kunz
- Institute of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
33
|
Triantis V, Bode L, van Neerven RJJ. Immunological Effects of Human Milk Oligosaccharides. Front Pediatr 2018; 6:190. [PMID: 30013961 PMCID: PMC6036705 DOI: 10.3389/fped.2018.00190] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/08/2018] [Indexed: 12/15/2022] Open
Abstract
Human milk oligosaccharides (HMOs) comprise a group of structurally complex, unconjugated glycans that are highly abundant in human milk. HMOs are minimally digested in the gastrointestinal tract and reach the colon intact, where they shape the microbiota. A small fraction of HMOs is absorbed, reaches the systemic circulation, and is excreted in urine. HMOs can bind to cell surface receptors expressed on epithelial cells and cells of the immune system and thus modulate neonatal immunity in the infant gut, and possibly also sites throughout the body. In addition, they have been shown to act as soluble decoy receptors to block the attachment of various microbial pathogens to cells. This review summarizes the current knowledge of the effects HMOs can have on infections, allergies, auto-immune diseases and inflammation, and will focus on the role of HMOs in altering immune responses through binding to immune-related receptors.
Collapse
Affiliation(s)
| | - Lars Bode
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
| | - R J Joost van Neerven
- FrieslandCampina, Amersfoort, Netherlands.,Wageningen University and Research, Cell Biology and Immunology, Wageningen, Netherlands
| |
Collapse
|
34
|
Williams JE, Price WJ, Shafii B, Yahvah KM, Bode L, McGuire MA, McGuire MK. Relationships Among Microbial Communities, Maternal Cells, Oligosaccharides, and Macronutrients in Human Milk. J Hum Lact 2017; 33:540-551. [PMID: 28609134 DOI: 10.1177/0890334417709433] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Human milk provides all essential nutrients necessary for early life and is rich in nonnutrients, maternally derived (host) cells, and bacteria, but almost nothing is known about the interplay among these components. Research aim: The primary objective of this research was to characterize relationships among macronutrients, maternal cells, and bacteria in milk. METHODS Milk samples were collected from 16 women and analyzed for protein, lipid, fatty acid, lactose, and human milk oligosaccharide concentrations. Concentrations of maternal cells were determined using microscopy, and somatic cell counts were enumerated. Microbial ecologies were characterized using culture-independent methods. RESULTS Absolute and relative concentrations of maternal cells were mostly consistent within each woman as were relative abundances of bacterial genera, and there were many apparent relationships between these factors. For instance, relative abundance of Serratia was negatively associated with somatic cell counts ( r = -.47, p < .0001) and neutrophil concentration ( r = -.38, p < .0006). Concentrations of several oligosaccharides were correlated with maternally derived cell types as well as somatic cell counts; for example, lacto-N-tetraose and lacto-N-neotetraose were inversely correlated with somatic cell counts ( r = -.64, p = .0082; r = -.52, p = .0387, respectively), and relative abundance of Staphylococcus was positively associated with total oligosaccharide concentration ( r = .69, p = .0034). Complex relationships between milk nutrients and bacterial community profile, maternal cells, and milk oligosaccharides were also apparent. CONCLUSION These data support the possibility that profiles of maternally derived cells, nutrient concentrations, and the microbiome of human milk might be interrelated.
Collapse
Affiliation(s)
- Janet E Williams
- 1 Department of Animal and Veterinary Science, University of Idaho, Moscow, ID, USA
| | - William J Price
- 2 Statistical Programs, University of Idaho, Moscow, ID, USA
| | - Bahman Shafii
- 2 Statistical Programs, University of Idaho, Moscow, ID, USA
| | - Katherine M Yahvah
- 1 Department of Animal and Veterinary Science, University of Idaho, Moscow, ID, USA
| | - Lars Bode
- 3 Department of Pediatrics, Mother-Milk-Infant Center of Research Excellence (MoMI CoRE), University of California, San Diego, La Jolla, CA, USA
| | - Mark A McGuire
- 1 Department of Animal and Veterinary Science, University of Idaho, Moscow, ID, USA
| | - Michelle K McGuire
- 4 Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (LRF MoMI CoRE), School of Medicine, University of California, San Diego, La Jolla, CA, USA.,5 School of Biological Sciences, Washington State University, Pullman, WA, USA
| |
Collapse
|
35
|
Dai Z, Lyu W, Xie M, Yuan Q, Ye H, Hu B, Zhou L, Zeng X. Effects of α-Galactooligosaccharides from Chickpeas on High-Fat-Diet-Induced Metabolic Syndrome in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:3160-3166. [PMID: 28362090 DOI: 10.1021/acs.jafc.7b00489] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The gut microbiota has the ability to modulate host energy homeostasis, which may regulate metabolic disorders. Functional oligosaccharide may positively regulate the intestinal microbiota. Therefore, effects of α-galactooligosaccharides (α-GOS) from chickpea on high-fat-diet (HFD)-induced metabolic syndrome and gut bacterial dysbiosis were investigated. After 6 weeks of intervention, HFD led to significant increases in levels of blood glucose, total cholesterol, triglyceride, glycated serum protein, high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol of mice compared to normal-chow-fed mice. Meanwhile, all of the α-GOS-treated groups significantly decreased above parameters compared to the HFD group. HFD could significantly decrease the content of all bacteria, especially Bacteroides (9.82 ± 0.09 versus 10.3 ± 0.10; p < 0.05) and Lactobacillus (6.67 ± 0.18 versus 7.30 ± 0.24; p < 0.05), and a decrease in the production of short-chain fatty acids was also observed. Treatment with α-GOS significantly increased the number of Bifidobacterium (6.07 ± 0.23 of the low-dose treatment versus 5.65 ± 0.20 of the HFD group) and Lactobacillus (7.22 ± 0.16 of the low-dose treatment). It also significantly promoted the secretion of propionic and butyric acids. These results indicate that α-GOS from chickpeas may affect the metabolic disorders and gut bacterial ecosystem in a positive way.
Collapse
Affiliation(s)
- Zhuqing Dai
- College of Food Science and Technology, Nanjing Agricultural University , Nanjing, Jiangsu 210095, People's Republic of China
| | - Wanyong Lyu
- Nutrition and Food Branch of China Association of Gerontology and Geriatrics , Beijing 100050, People's Republic of China
| | - Minhao Xie
- College of Food Science and Technology, Nanjing Agricultural University , Nanjing, Jiangsu 210095, People's Republic of China
| | - Qingxia Yuan
- College of Food Science and Technology, Nanjing Agricultural University , Nanjing, Jiangsu 210095, People's Republic of China
| | - Hong Ye
- College of Food Science and Technology, Nanjing Agricultural University , Nanjing, Jiangsu 210095, People's Republic of China
| | - Bing Hu
- College of Food Science and Technology, Nanjing Agricultural University , Nanjing, Jiangsu 210095, People's Republic of China
| | - Li Zhou
- College of Food Science and Technology, Nanjing Agricultural University , Nanjing, Jiangsu 210095, People's Republic of China
| | - Xiaoxiong Zeng
- College of Food Science and Technology, Nanjing Agricultural University , Nanjing, Jiangsu 210095, People's Republic of China
| |
Collapse
|
36
|
Lourenço AG, Komesu MC, Duarte G, Del Ciampo LA, Mussi-Pinhata MM, Yamamoto AY. High Levels of Chemokine C-C Motif Ligand 20 in Human Milk and Its Production by Oral Keratinocytes. Breastfeed Med 2017; 12:116-121. [PMID: 27912038 DOI: 10.1089/bfm.2016.0067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Chemokine C-C motif ligand 20 (CCL20) is implicated in the formation and function of mucosal lymphoid tissues. Although CCL20 is secreted by many normal human tissues, no studies have evaluated the presence of CCL20 in human milk or its production by oral keratinocytes stimulated by human milk. OBJECTIVE To evaluate the presence of CCL20 in breast milk and verify CCL20 secretion in vitro by oral keratinocytes stimulated with human and bovine milk, as well as its possible association with breast milk lactoferrin levels. MATERIALS AND METHODS The levels of CCL20 and lactoferrin were measured by enzyme-linked immunosorbent assay in human milk at three different stages of maturation from 74 healthy breastfeeding mothers. In vitro, oral keratinocytes were stimulated with human and bovine milk, and CCL20 was measured in their supernatant. RESULTS High concentrations of CCL20 were detected in the human breast milk samples obtained during the first week (1,777.07 pg/mL) and second week postpartum (1,523.44 pg/mL), with a significantly low concentration in samples at 3-6 weeks postpartum (238.42 pg/mL; p < 0.0001). Human breast milk at different weeks postpartum stimulated higher CCL20 secretion by oral keratinocytes compared with bovine milk (p < 0.05). Such stimulation had no association with breast milk lactoferrin concentration. CONCLUSION CCl20 is present at high levels in human milk, predominantly in the first and second week postpartum, but at significantly lower levels at 3-6 weeks postpartum. Human milk is capable of stimulating CCL20 secretion by oral keratinocytes, and this induction had no association with breast milk lactoferrin concentration.
Collapse
Affiliation(s)
- Alan G Lourenço
- 1 Department of Pediatrics, Ribeirão Preto School of Medicine, University of São Paulo , São Paulo, Brazil
| | - Marilena C Komesu
- 2 Department of Morphology, Physiology and Basic Pathology, Ribeirão Preto School of Dentistry, University of São Paulo , São Paulo, Brazil
| | - Geraldo Duarte
- 3 Department of Gynecology and Obstetrics, Ribeirão Preto School of Medicine, University of São Paulo , São Paulo, Brazil
| | - Luiz A Del Ciampo
- 1 Department of Pediatrics, Ribeirão Preto School of Medicine, University of São Paulo , São Paulo, Brazil
| | - Marisa M Mussi-Pinhata
- 1 Department of Pediatrics, Ribeirão Preto School of Medicine, University of São Paulo , São Paulo, Brazil
| | - Aparecida Y Yamamoto
- 1 Department of Pediatrics, Ribeirão Preto School of Medicine, University of São Paulo , São Paulo, Brazil
| |
Collapse
|
37
|
Donovan SM, Comstock SS. Human Milk Oligosaccharides Influence Neonatal Mucosal and Systemic Immunity. ANNALS OF NUTRITION AND METABOLISM 2017; 69 Suppl 2:42-51. [PMID: 28103609 DOI: 10.1159/000452818] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The immune system of the infant is functionally immature and naïve. Human milk contains bioactive proteins, lipids, and carbohydrates that protect the newborn and stimulate innate and adaptive immune development. This review will focus on the role human milk oligosaccharides (HMO) play in neonatal gastrointestinal and systemic immune development and function. For the past decade, intense research has been directed at defining the complexity of oligosaccharides in the milk of many species and is beginning to delineate their diverse functions. These studies have shown that human milk contains a higher concentration as well as a greater structural diversity and degree of fucosylation than the milk oligosaccharides in other species, particularly bovine milk from which many infant formulae are produced. The commercial availability of large quantities of certain HMO has furthered our understanding of the functions of specific HMO, which include protecting the infant from pathogenic infections, facilitating the establishment of the gut microbiota, promoting intestinal development, and stimulating immune maturation. Many of these actions are exerted through carbohydrate-carbohydrate interactions with pathogens or host cells. Two HMOs, 2'-fucosyllactose (2'FL) and lacto-N-neotetraose (LNnT), have recently been added to infant formula. Although this is a first step in narrowing the compositional gap between human milk and infant formula, it is unclear whether 1 or 2 HMO will recapitulate the complexity of actions exerted by the complex mixture of HMO ingested by breastfed infants. Thus, as more HMO become commercially available, either isolated from bovine milk or chemically or microbially synthesized, it is anticipated that more oligosaccharides will be added to infant formula either alone or in combination with other prebiotics.
Collapse
Affiliation(s)
- Sharon M Donovan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL, USA
| | | |
Collapse
|
38
|
The Role of Oligosaccharides in Host-Microbial Interactions for Human Health. J Clin Gastroenterol 2016; 50 Suppl 2, Proceedings from the 8th Probiotics, Prebiotics & New Foods for Microbiota and Human Health meeting held in Rome, Italy on September 13-15, 2015:S131-S132. [PMID: 27741156 DOI: 10.1097/mcg.0000000000000694] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Milk oligosaccharides have many associated bioactivities which can contribute to human health and offer protective properties to the host. Such bioactivities include anti-infective properties whereby oligosaccharides interact with bacterial cells and prevent adhesion to the host and subsequent colonization. Milk oligosaccharides have also been shown to alter the glycosylation of intestinal cells, leading to a reduction in pathogenic colonization. In addition, these sugars promote adhesion of commensal bacterial strains to host cells as well as possessing the ability to alter mucin expression in intestinal cells and improve barrier function. The ability of milk oligosaccharides to alter the transcriptome of both commensal bacterial strains and intestinal epithelial cells has also been revealed, indicating the potential of many cell types to detect the presence of milk oligosaccharides and respond accordingly at the genetic level. Interestingly, domestic animal milk may provide a bioactive source of oligosaccharides for formula supplementation with the aim of emulating the gold standard that is human milk. Overall, this review highlights the ability of milk oligosaccharides to promote health in a variety of ways, for example, through direct bacterial interactions, immunomodulatory activities, promotion of gut barrier function, and induction of protective transcriptional responses.
Collapse
|
39
|
Characterization of two novel sialyl N-acetyllactosaminyl nucleotides separated from ovine colostrum. Glycoconj J 2016; 33:789-96. [PMID: 27206414 DOI: 10.1007/s10719-016-9672-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/15/2016] [Accepted: 05/02/2016] [Indexed: 01/31/2023]
Abstract
The milk/colostrum of some mammalian species is known to contain sugar nucleotides including uridine diphosphate (UDP) oligosaccharides in addition to lactose and milk oligosaccharides, but the detailed structures of these UDP oligosaccharides have not so far been clarified. In this study we isolated two UDP-sialyl N-acetyllactosamines from ovine colostrum and characterized them using (1)H-NMR and MALDI-TOFMS spectroscopies. Their structures were found to be Neu5Gc(α2-3)Gal(β1-4)GlcNAcα1-UDP and Neu5Gc(α2-6)Gal(β1-4)GlcNAcα1-UDP.
Collapse
|
40
|
Abstract
Toll-like receptor (TLR) signaling is central to innate immunity. Aberrant expression of TLRs is found in neonatal inflammatory diseases. Several bioactive components of human milk modulate TLR expression and signaling pathways, including soluble toll-like receptors (sTLRs), soluble cluster of differentiation (sCD) 14, glycoproteins, small peptides, and oligosaccharides. Some milk components, such as sialyl (α2,3) lactose and lacto-N-fucopentaose III, are reported to increase TLR signaling; under some circumstances this might contribute toward immunologic balance. Human milk on the whole is strongly anti-inflammatory, and contains abundant components that depress TLR signaling pathways: sTLR2 and sCD14 inhibit TLR2 signaling; sCD14, lactadherin, lactoferrin, and 2'-fucosyllactose attenuate TLR4 signaling; 3'-galactosyllactose inhibits TLR3 signaling, and β-defensin 2 inhibits TLR7 signaling. Feeding human milk to neonates decreases their risk of sepsis and necrotizing enterocolitis. Thus, the TLR regulatory components found in human milk hold promise as benign oral prophylactic and therapeutic treatments for the many gastrointestinal inflammatory disorders mediated by abnormal TLR signaling.
Collapse
Affiliation(s)
- YingYing He
- Program in Glycobiology, Department of Biology, Boston College, Chestnut Hill, MA
| | - Nathan T Lawlor
- Program in Glycobiology, Department of Biology, Boston College, Chestnut Hill, MA
| | - David S Newburg
- Program in Glycobiology, Department of Biology, Boston College, Chestnut Hill, MA
| |
Collapse
|
41
|
Altmann K, Wutkowski A, Kämpfer S, Klempt M, Lorenzen PC, Clawin-Rädecker I. Comparison of the efficiency of different NF membranes for the enrichment of milk oligosaccharides from bovine milk. Eur Food Res Technol 2015. [DOI: 10.1007/s00217-015-2505-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
42
|
Hermosilla C, Stamm I, Menge C, Taubert A. Suitable in vitro culture of Eimeria bovis meront II stages in bovine colonic epithelial cells and parasite-induced upregulation of CXCL10 and GM-CSF gene transcription. Parasitol Res 2015; 114:3125-36. [PMID: 25982572 DOI: 10.1007/s00436-015-4531-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/06/2015] [Indexed: 12/11/2022]
Abstract
We here established a suitable in vitro cell culture system based on bovine colonic epithelial cells (BCEC) for the development of Eimeria bovis merozoites I and the characterization of early parasite-induced innate epithelial host cell reactions as gene transcription of proinflammatory molecules. Both primary and permanent BCEC (BCEC (rim) and BCEC(perm)) were suitable for E. bovis merozoite I invasion and subsequent development of meronts II leading to the release of viable merozoites II. E. bovis merozoite II failed to develop any further neither into gamont nor oocyst stages in BCEC in vitro. E. bovis merozoite I induced innate epithelial host cell reactions at the level of CXC/CCL chemokines (CXCL1, CXCL8, CXCL10, CCL2), IL-6, and GM-CSF gene transcription. Overall, both BCEC types were activated by merozoite I infections since they showed significantly enhanced gene transcript levels of the immunomodulatory molecules CXCL10 and GM-CSF. However, gene transcription profiles of BCEC(prim) and BCEC(perm) revealed different reaction patterns in response to merozoite I infection with regard to quality and kinetics of chemokine/cytokine gene transcription. Although both BCEC types equally showed most prominent responses for CXCL10 and GM-CSF, the induction of CXCL1, CXCL8, CCL2, and IL-6 gene transcripts varied qualitatively and quantitatively. Our results demonstrate that BCEC seem capable to respond to E. bovis merozoite I infection by the upregulation of CXCL10 and GM-CSF gene transcription and therefore probably contribute to host innate effector mechanisms against E. bovis.
Collapse
Affiliation(s)
- Carlos Hermosilla
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, Giessen, Germany,
| | | | | | | |
Collapse
|
43
|
Donovan SM, Wang M, Monaco MH, Martin CR, Davidson LA, Ivanov I, Chapkin RS. Noninvasive molecular fingerprinting of host-microbiome interactions in neonates. FEBS Lett 2014; 588:4112-9. [PMID: 25042036 DOI: 10.1016/j.febslet.2014.07.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 07/09/2014] [Accepted: 07/10/2014] [Indexed: 01/12/2023]
Abstract
The early postnatal period is a critical window for intestinal and immune maturation. Intestinal development and microbiome diversity and composition differ between breast- (BF) and formula-fed (FF) infants. Mechanistic examination into host-microbe relationships in healthy infants has been hindered by ethical constraints surrounding tissue biopsies. Thus, a statistically rigorous analytical framework to simultaneously examine both host and microbial responses to dietary/environmental factors using exfoliated intestinal epithelial cells was developed. Differential expression of ∼1200 genes, including genes regulating intestinal proliferation, differentiation and barrier function, was observed between BF and FF term infants. Canonical correlation analysis uncovered a relationship between microbiome virulence genes and host immunity and defense genes. Lastly, exfoliated cells from preterm and term infants were compared. Pathways associated with immune cell function and inflammation were up-regulated in preterm, whereas cell growth-related genes were up-regulated in the term infants. Thus, coordinate measurement of the transcriptomes of exfoliated epithelial cells and microbiome allows inquiry into mutualistic host-microbe interactions in the infant, which can be used to prospectively study gut development or, retrospectively, to identify potential triggers of disease in banked samples.
Collapse
Affiliation(s)
- Sharon M Donovan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL 61801, USA.
| | - Mei Wang
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL 61801, USA
| | - Marcia H Monaco
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL 61801, USA
| | - Camilia R Martin
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Laurie A Davidson
- Department of Nutrition & Food Science and Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX 77843-2253, USA; Center for Translational Environmental Health Research, Texas A&M University, College Station, TX 77843-2253, USA
| | - Ivan Ivanov
- Department of Nutrition & Food Science and Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX 77843-2253, USA; Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-2253, USA; Center for Translational Environmental Health Research, Texas A&M University, College Station, TX 77843-2253, USA
| | - Robert S Chapkin
- Department of Nutrition & Food Science and Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX 77843-2253, USA; Center for Translational Environmental Health Research, Texas A&M University, College Station, TX 77843-2253, USA
| |
Collapse
|
44
|
Capitán-Cañadas F, Ortega-González M, Guadix E, Zarzuelo A, Suárez MD, de Medina FS, Martínez-Augustin O. Prebiotic oligosaccharides directly modulate proinflammatory cytokine production in monocytes via activation of TLR4. Mol Nutr Food Res 2013; 58:1098-110. [PMID: 24549946 DOI: 10.1002/mnfr.201300497] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 09/10/2013] [Accepted: 10/12/2013] [Indexed: 11/06/2022]
Abstract
SCOPE Prebiotic oligosaccharides are currently used in a variety of clinical settings for their effects on intestinal microbiota. Here, we have examined the direct, microbiota independent, effects of prebiotics on monocytes and T lymphocytes in vitro. METHODS AND RESULTS Prebiotics generally evoked cytokine secretion (TNF-α, IL-6, and IL-10) by mouse splenocytes but inhibited LPS -induced IFN-γ and IL-17 release. Inulin was found to enhance LPS-induced IL-10 secretion. Splenocytes from TLR4(-/-) (where TLR is Toll-like receptor) mice showed a markedly depressed response. Conversely, in both basal and LPS-stimulated conditions, prebiotic inhibition of IFN-γ levels was preserved. These results suggested a predominant effect on monocytes via TLR4 ligation and possible inhibition of T cells. Hence, we studied the modulation of primary rat monocytes and T lymphocytes, focusing on fructooligosaccharides (FOS) and inulin. In monocytes, FOS and inulin induced TNF-α, growth-regulated oncogene α, and IL-10, but not IL-1β release. The NF-κB inhibitor Bay 11-7082 fully prevented these effects. Pharmacological evidence also indicated a significant involvement of mitogen-activated protein kinase and phosphatidylinositol-3-kinase. There was little effect on T cells. FOS and inulin also generally increased TNF-α, IL-1β, and IL-10, but not IL-8, in human peripheral blood monocytes. CONCLUSION We conclude that prebiotics may act as TLR4 ligands or as indirect TLR4 modulators to upregulate cytokine secretion in monocytes.
Collapse
Affiliation(s)
- Fermín Capitán-Cañadas
- Department of Biochemistry and Molecular Biology II, CIBERehd, School of Pharmacy, University of Granada, Granada, Spain
| | | | | | | | | | | | | |
Collapse
|