1
|
Kapoor B, Biswas P, Gulati M, Rani P, Gupta R. Gut microbiome and Alzheimer's disease: What we know and what remains to be explored. Ageing Res Rev 2024; 102:102570. [PMID: 39486524 DOI: 10.1016/j.arr.2024.102570] [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: 06/18/2024] [Revised: 10/26/2024] [Accepted: 10/26/2024] [Indexed: 11/04/2024]
Abstract
With advancement in human microbiome research, an increasing number of scientific evidences have endorsed the key role of gut microbiota in the pathogenesis of Alzheimer disease. Microbiome dysbiosis, characterized by altered diversity and composition, as well as rise of pathobionts influence not only various gut disorder but also central nervous system disorders such as AD. On the basis of accumulated evidences of past few years now it is quite clear that the gut microbiota can control the functions of the central nervous system (CNS) through the gut-brain axis, which provides a new prospective into the interactions between the gut and brain. The main focus of this review is on the molecular mechanism of the crosstalk between the gut microbiota and the brain through the gut-brain axis, and on the onset and development of neurological disorders triggered by the dysbiosis of gut microbiota. Due to microbiota dysbiosis the permeability of the gut and blood brain barrier is increased which may mediate or affect AD. Along with this, bacterial population of the gut microbiota can secrete amyloid proteins and lipopolysaccharides in a large quantity which may create a disturbance in the signaling pathways and the formation of proinflammatory cytokines associated with the pathogenesis of AD. These topics are followed by a critical analysis of potential intervention strategies targeting gut microbiota dysbiosis, including the use of probiotics, prebiotics, metabolites, diets and fecal microbiota transplantation. The main purpose of this review includes the summarization and discussion on the recent finding that may explain the role of the gut microbiota in the development of AD. Understanding of these fundamental mechanisms may provide a new insight into the novel therapeutic strategies for AD.
Collapse
Affiliation(s)
- Bhupinder Kapoor
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India.
| | - Pratim Biswas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, NSW 2007, Australia
| | - Pooja Rani
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Reena Gupta
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
2
|
Nie RZ, Luo HM, Liu YP, Wang SS, Hou YJ, Chen C, Wang H, Lv HL, Tao XY, Jing ZH, Zhang HK, Li PF. Food Functional Factors in Alzheimer's Disease Intervention: Current Research Progress. Nutrients 2024; 16:3998. [PMID: 39683391 DOI: 10.3390/nu16233998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease (AD) is a complex multifactorial neurodegenerative disease. With the escalating aging of the global population, the societal burden of this disease is increasing. Although drugs are available for the treatment of AD, their efficacy is limited and there remains no effective cure. Therefore, the identification of safe and effective prevention and treatment strategies is urgently needed. Functional factors in foods encompass a variety of natural and safe bioactive substances that show potential in the prevention and treatment of AD. However, current research focused on the use of these functional factors for the prevention and treatment of AD is in its initial stages, and a complete theoretical and application system remains to be determined. An increasing number of recent studies have found that functional factors such as polyphenols, polysaccharides, unsaturated fatty acids, melatonin, and caffeine have positive effects in delaying the progression of AD and improving cognitive function. For example, polyphenols exhibit antioxidant, anti-inflammatory, and neuroprotective effects, and polysaccharides promote neuronal growth and inhibit inflammation and oxidative stress. Additionally, unsaturated fatty acids inhibit Aβ production and Tau protein phosphorylation and reduce neuroinflammation, and melatonin has been shown to protect nerve cells and improve cognitive function by regulating mitochondrial homeostasis and autophagy. Caffeine has also been shown to inhibit inflammation and reduce neuronal damage. Future research should further explore the mechanisms of action of these functional factors and develop relevant functional foods or nutritional supplements to provide new strategies and support for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Rong-Zu Nie
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Huo-Min Luo
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Ya-Ping Liu
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Shuang-Shuang Wang
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Yan-Jie Hou
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Chen Chen
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Hang Wang
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Hui-Lin Lv
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Xing-Yue Tao
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Zhao-Hui Jing
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Hao-Kun Zhang
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Pei-Feng Li
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| |
Collapse
|
3
|
Sanchez-Gallardo R, Bottacini F, O’Neill IJ, Esteban-Torres M, Moore R, McAuliffe FM, Cotter PD, van Sinderen D. Selective human milk oligosaccharide utilization by members of the Bifidobacterium pseudocatenulatum taxon. Appl Environ Microbiol 2024; 90:e0064824. [PMID: 39315793 PMCID: PMC11497806 DOI: 10.1128/aem.00648-24] [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: 04/03/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
Human milk oligosaccharides (HMOs) are essentially unaffected by the digestive enzymes of the nursling and are known for their ability to enrich certain microbial species in the infant gut microbiota, in particular bifidobacteria. HMO metabolism has been studied in various bifidobacterial species such as B. breve, B. bifidum, and B. longum subsp. infantis. In the current study, we describe differential growth abilities elicited by twenty-three newly isolated Bifidobacterium pseudocatenulatum strains on particular HMOs, such as 2'-fucosyllactose (2'FL), 3-fucosyllactose (3FL), lacto-N-tetraose (LNT), and lacto-N-neotetraose (LNnT). Through gene-trait matching and comparative genome analysis, we identified genes involved in the degradation of fucosylated HMOs in this strain set, while we employed a transcriptomic approach to facilitate the identification and characterization of genes and associated enzymes involved in LNT metabolism by strain B. pseudocatenulatum MM0196. A total of 252 publicly available genomes of the B. pseudocatenulatum taxon were screened for homologs of the glycosyl hydrolases (GHs) identified here as being required for selected HMO metabolism. From this analysis, it is clear that all members of this species possess homologs of the genes involved in LNT degradation, while genes required for degradation of fucosylated HMOs are variably present.IMPORTANCEOur findings allow a better understanding of the complex interaction between Bifidobacterium and its host and provide a roadmap toward future applications of B. pseudocatenulatum as a probiotic with a focus on infant health. Furthermore, our investigations have generated information on the role of HMOs in shaping the infant gut microbiota, thus also facilitating applications of HMOs in infant nutrition, with potential extension into the mature or adult gut microbiota. Supplementation of HMOs is known to result in the modulation of bacterial communities toward a higher relative abundance of bifidobacteria, which in turn enforces their ability to modulate particular immune functions and strengthen the intestinal barrier. This work may therefore inspire future studies to improve the formulation of neonatal nutritional products, aimed at facilitating the development of a healthy digestive and immune system and reducing the differences in gut microbiota composition observed between breastfed and formula-fed babies or full-term and preterm infants.
Collapse
Affiliation(s)
- Rocio Sanchez-Gallardo
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Francesca Bottacini
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Biological Sciences and ADAPT, Munster Technological University, Cork, Ireland
| | - Ian J. O’Neill
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Maria Esteban-Torres
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Rebecca Moore
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Ireland
| | - Fionnuala M. McAuliffe
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Ireland
| | - Paul D. Cotter
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre Moorepark, Cork, Ireland
| | - Douwe van Sinderen
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
4
|
Suárez-Martínez C, Santaella-Pascual M, Yagüe-Guirao G, García-Marcos L, Ros G, Martínez-Graciá C. The Early Appearance of Asthma and Its Relationship with Gut Microbiota: A Narrative Review. Microorganisms 2024; 12:1471. [PMID: 39065238 PMCID: PMC11278858 DOI: 10.3390/microorganisms12071471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Asthma is, worldwide, the most frequent non-communicable disease affecting both children and adults, with high morbidity and relatively low mortality, compared to other chronic diseases. In recent decades, the prevalence of asthma has increased in the pediatric population, and, in general, the risk of developing asthma and asthma-like symptoms is higher in children during the first years of life. The "gut-lung axis" concept explains how the gut microbiota influences lung immune function, acting both directly, by stimulating the innate immune system, and indirectly, through the metabolites it generates. Thus, the process of intestinal microbial colonization of the newborn is crucial for his/her future health, and the alterations that might generate dysbiosis during the first 100 days of life are most influential in promoting hypersensitivity diseases. That is why this period is termed the "critical window". This paper reviews the published evidence on the numerous factors that can act by modifying the profile of the intestinal microbiota of the infant, thereby promoting or inhibiting the risk of asthma later in life. The following factors are specifically addressed in depth here: diet during pregnancy, maternal adherence to a Mediterranean diet, mode of delivery, exposure to antibiotics, and type of infant feeding during the first three months of life.
Collapse
Affiliation(s)
- Clara Suárez-Martínez
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain; (C.S.-M.); (G.Y.-G.); (G.R.)
- Food Science and Nutrition Department, Veterinary Faculty, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, 30100 Murcia, Spain
| | - Marina Santaella-Pascual
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain; (C.S.-M.); (G.Y.-G.); (G.R.)
- Food Science and Nutrition Department, Veterinary Faculty, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, 30100 Murcia, Spain
| | - Genoveva Yagüe-Guirao
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain; (C.S.-M.); (G.Y.-G.); (G.R.)
- Microbiology Service, Virgen de La Arrixaca University Clinical Hospital, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, 30120 Murcia, Spain
| | - Luis García-Marcos
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain; (C.S.-M.); (G.Y.-G.); (G.R.)
- Pediatric Allergy and Pulmonology Units, Virgen de La Arrixaca University Clinical Hospital, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, 30120 Murcia, Spain
- Network of Asthma and Adverse and Allergic Reactions (ARADyAL), 28029 Madrid, Spain
| | - Gaspar Ros
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain; (C.S.-M.); (G.Y.-G.); (G.R.)
- Food Science and Nutrition Department, Veterinary Faculty, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, 30100 Murcia, Spain
| | - Carmen Martínez-Graciá
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain; (C.S.-M.); (G.Y.-G.); (G.R.)
- Food Science and Nutrition Department, Veterinary Faculty, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, 30100 Murcia, Spain
| |
Collapse
|
5
|
Korpela K, Hurley S, Ford SA, Franklin R, Byrne S, Lunjani N, Forde B, Neogi U, Venter C, Walter J, Hourihane J, O'Mahony L. Association between gut microbiota development and allergy in infants born during pandemic-related social distancing restrictions. Allergy 2024; 79:1938-1951. [PMID: 38419554 DOI: 10.1111/all.16069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/05/2024] [Accepted: 02/06/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Several hypotheses link reduced microbial exposure to increased prevalence of allergies. Here we capitalize on the opportunity to study a cohort of infants (CORAL), raised during COVID-19 associated social distancing measures, to identify the environmental exposures and dietary factors that contribute to early life microbiota development and to examine their associations with allergic outcomes. METHODS Fecal samples were sequenced from infants at 6 (n = 351) and repeated at 12 (n = 343) months, using 16S sequencing. Published 16S data from pre-pandemic cohorts were included for microbiota comparisons. Online questionnaires collected epidemiological information on home environment, healthcare utilization, infant health, allergic diseases, and diet. Skin prick testing (SPT) was performed at 12 (n = 343) and 24 (n = 320) months of age, accompanied by atopic dermatitis and food allergy assessments. RESULTS The relative abundance of bifidobacteria was higher, while environmentally transmitted bacteria such as Clostridia was lower in CORAL infants compared to previous cohorts. The abundance of multiple Clostridia taxa correlated with a microbial exposure index. Plant based foods during weaning positively impacted microbiota development. Bifidobacteria levels at 6 months of age, and relative abundance of butyrate producers at 12 months of age, were negatively associated with AD and SPT positivity. The prevalence of allergen sensitization, food allergy, and AD did not increase over pre-pandemic levels. CONCLUSIONS Environmental exposures and dietary components significantly impact microbiota community assembly. Our results also suggest that vertically transmitted bacteria and appropriate dietary supports may be more important than exposure to environmental microbes alone for protection against allergic diseases in infancy.
Collapse
Affiliation(s)
- Katri Korpela
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Sadhbh Hurley
- Paediatrics and Child Health, Royal College of Surgeons in Ireland, Dublin, Ireland
- Children's Health Ireland, Dublin, Ireland
| | | | - Ruth Franklin
- Paediatrics and Child Health, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Susan Byrne
- Paediatrics and Child Health, Royal College of Surgeons in Ireland, Dublin, Ireland
- Children's Health Ireland, Dublin, Ireland
| | | | - Brian Forde
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Ujjwal Neogi
- The Systems Virology Lab, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Carina Venter
- Section of Allergy & Immunology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jens Walter
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| | - Jonathan Hourihane
- Paediatrics and Child Health, Royal College of Surgeons in Ireland, Dublin, Ireland
- Children's Health Ireland, Dublin, Ireland
| | - Liam O'Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| |
Collapse
|
6
|
Chen YJ, Sui X, Wang Y, Zhao ZH, Han TH, Liu YJ, Zhang JN, Zhou P, Yang K, Ye ZH. Preparation, structural characterization, biological activity, and nutritional applications of oligosaccharides. Food Chem X 2024; 22:101289. [PMID: 38544933 PMCID: PMC10966145 DOI: 10.1016/j.fochx.2024.101289] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 11/11/2024] Open
Abstract
Oligosaccharides are low-molecular-weight carbohydrates between monosaccharides and polysaccharides. They can be extracted directly from natural products by physicochemical methods or obtained by chemical synthesis or enzymatic reaction. Oligosaccharides have important physicochemical and physiological properties. Their research and production involve many disciplines such as medicine, chemical industry, and biology. Functional oligosaccharides, as an excellent functional food base, can be used as dietary fibrer and prebiotics to enrich the diet; improve the microecology of the gut; exert antitumour, anti-inflammatory, antioxidant, and lipid-lowering properties. Therefore, the industrial applications of oligosaccharides have increased rapidly in the past few years. It has great prospects in the field of food and medicinal chemistry. This review summarized the preparation, structural features and biological activities of oligosaccharides, with particular emphasis on the application of functional oligosaccharides in the food industry and human nutritional health. It aims to inform further research and development of oligosaccharides and food chemistry.
Collapse
Affiliation(s)
- Ya-jing Chen
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Xin Sui
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yue Wang
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Zhi-hui Zhao
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Tao-hong Han
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Yi-jun Liu
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Jia-ning Zhang
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Ping Zhou
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49, Huayuan North Road, Haidian District, Beijing 100191, China
| | - Ke Yang
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou 310018, China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Beijing 100700, China
| | - Zhi-hong Ye
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou 310018, China
| |
Collapse
|
7
|
Hick E, Suárez M, Rey A, Mantecón L, Fernández N, Solís G, Gueimonde M, Arboleya S. Personalized Nutrition with Banked Human Milk for Early Gut Microbiota Development: In Pursuit of the Perfect Match. Nutrients 2024; 16:1976. [PMID: 38999725 PMCID: PMC11243202 DOI: 10.3390/nu16131976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
The correct initial colonization and establishment of the gut microbiota during the early stages of life is a key step, with long-lasting consequences throughout the entire lifespan of the individual. This process is affected by several perinatal factors; among them, feeding mode is known to have a critical role. Breastfeeding is the optimal nutrition for neonates; however, it is not always possible, especially in cases of prematurity or early pathology. In such cases, most commonly babies are fed with infant formulas in spite of the official nutritional and health international organizations' recommendation on the use of donated human milk through milk banks for these cases. However, donated human milk still does not totally match maternal milk in terms of infant growth and gut microbiota development. The present review summarizes the practices of milk banks and hospitals regarding donated human milk, its safety and quality, and the health outcomes in infants fed with donated human milk. Additionally, we explore different alternatives to customize pasteurized donated human milk with the aim of finding the perfect match between each baby and banked milk for promoting the establishment of a beneficial gut microbiota from the early stages of life.
Collapse
Affiliation(s)
- Emilia Hick
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain
| | - Marta Suárez
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Alejandra Rey
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain
| | - Laura Mantecón
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Nuria Fernández
- Pediatrics Service, University Hospital of Cabueñes (CAB-SESPA), 33394 Gijón, Spain
| | - Gonzalo Solís
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
8
|
Ferreres-Serafini L, Martín-Orúe SM, Sadurní M, Jiménez J, Moreno-Muñoz JA, Castillejos L. Supplementing infant milk formula with a multi-strain synbiotic and osteopontin enhances colonic microbial colonization and modifies jejunal gene expression in lactating piglets. Food Funct 2024; 15:6536-6552. [PMID: 38807503 DOI: 10.1039/d4fo00489b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
A total of ninety-six weaned piglets were assigned to four dietary treatments in a 2 × 2 design. The treatments included: a standard milk formula (CTR); CTR + probiotics (6.4 × 108 cfu L-1Bifidobacterium longum subsp. infantis CECT 7210 and 1.1 × 108 cfu L-1Lactobacillus rhamnosus NH001) + prebiotics (galacto-oligosaccharides 4.36 g L-1 and human-milk-oligosaccharide 0.54 g L-1) (SYN); CTR + osteopontin (0.43 g L-1) (OPN); and CTR + SYN + OPN (CON). Daily records including feed intake, body weight, and clinical signs, were maintained throughout the 15-day trial. At the end of the study samples from blood, digestive content, and gut tissues were collected to determine serum TNF-α, intestinal fermentative activity (SCFA and ammonia), colonic microbiota (16S rRNA Illumina-MiSeq), histomorphology, and jejunal gene expression (Open-Array). No statistical differences were found in weight gain; however, the animals supplemented with osteopontin exhibited higher feed intake. In terms of clinical signs, synbiotic supplementation led to a shorter duration of diarrhoea episodes. Regarding gut health, the sequenced faecal microbiota revealed better control of potentially dysbiotic bacteria with the CON diet at day 15. In the colon compartment, a significant increase in SCFA concentration, a decrease in ammonia concentration, and a significant decrease in intraepithelial lymphocyte counts were particularly observed in CON animals. The supplemented diets were also associated with modified jejunal gene expression. The synbiotic combination was characterized by the upregulation of genes related to intestinal maturation (ALPI, SI) and nutrient transport (SLC13A1, SLC15A1, SLC5A1, SLC7A8), and the downregulation of genes related to the response to pathogens (GBP1, IDO, TLR4) or the inflammatory response (IDO, IL-1β, TGF-β1). Osteopontin promoted the upregulation of a digestive function gene (GCG). Correlational analysis between the microbiota population and various intestinal environmental factors (SCFA concentration, histology, and gene expression) proposes mechanisms of communication between the gut microbiota and the host. In summary, these results suggest an improvement in the colonic colonization process and a better modulation of the immune response when milk formula is supplemented with the tested synbiotic combined with osteopontin, benefiting from a synergistic effect.
Collapse
Affiliation(s)
- Laia Ferreres-Serafini
- Animal Nutrition and Welfare Service, Department of Animal and Food Science, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
| | - Susana Mª Martín-Orúe
- Animal Nutrition and Welfare Service, Department of Animal and Food Science, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
| | - Meritxell Sadurní
- Animal Nutrition and Welfare Service, Department of Animal and Food Science, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
| | - Jesús Jiménez
- Laboratorios Ordesa S.L., Parc Científic de Barcelona, C/Baldiri Reixac 15-21, 08028 Barcelona, Spain.
| | - José Antonio Moreno-Muñoz
- Laboratorios Ordesa S.L., Parc Científic de Barcelona, C/Baldiri Reixac 15-21, 08028 Barcelona, Spain.
| | - Lorena Castillejos
- Animal Nutrition and Welfare Service, Department of Animal and Food Science, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
| |
Collapse
|
9
|
Tran LC, Marousez L, Micours E, De Lamballerie M, Thys L, Gottrand F, Ley D, Lesage J, Titécat M. High hydrostatic pressure is similar to Holder pasteurization in preserving donor milk antimicrobial activity. Pediatr Res 2024; 95:1749-1753. [PMID: 38280953 DOI: 10.1038/s41390-024-03022-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 12/22/2023] [Accepted: 12/29/2023] [Indexed: 01/29/2024]
Abstract
BACKGROUND The microbiological safety of donor milk (DM) is commonly ensured by Holder pasteurization (HoP, 62.5 °C for 30 min) in human milk banks despite its detrimental effects on bioactive factors. We compared the antimicrobial properties of DM after Holder pasteurization treatment or High Hydrostatic Pressure processing (HHP, 350 MPa at 38 °C), a non-thermal substitute for DM sterilization. METHODS We assessed lactoferrin and lysozyme concentrations in raw, HHP- and HoP-treated pools of DM (n = 8). The impact of both treatments was evaluated on the growth of Escherichia coli and Group B Streptococcus in comparison with control media (n = 4). We also addressed the effect of storage of HHP treated DM over a 6-month period (n = 15). RESULTS HHP milk demonstrated similar concentrations of lactoferrin compared with raw milk, while it was significantly decreased by HoP. Lysozyme concentrations remained stable regardless of the condition. Although a bacteriostatic effect was observed against Escherichia coli at early timepoints, a sharp bactericidal effect was observed against Group B Streptococcus. Unlike HoP, these results were significant for HHP compared to controls. Stored DM was well and safely preserved by HHP. CONCLUSION Our study demonstrates that this alternative sterilization method shows promise for use with DM in human milk banks. IMPACT Antimicrobial activity of donor milk after High Hydrostatic Pressure treatment has not been clearly evaluated. Donor milk lactoferrin is better preserved by High Hydrostatic Pressure than conventional Holder pasteurization, while lysozyme concentration is not affected by either treatment. As with Holder pasteurization, High Hydrostatic Pressure preserves donor milk bacteriostatic activity against E. coli in addition to bactericidal activity against Group B Streptococcus. Donor milk treated by High Hydrostatic Pressure can be stored safely for 6 months.
Collapse
Affiliation(s)
- Léa Chantal Tran
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France.
- Division of Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Jeanne de Flandre Children's Hospital, CHU Lille, F-59000, Lille, France.
| | - Lucie Marousez
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
| | - Edwina Micours
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
| | | | - Lou Thys
- Laboratory of Bacteriology, Institute of Microbiology, CHU Lille, Lille, France
| | - Frédéric Gottrand
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
- Division of Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Jeanne de Flandre Children's Hospital, CHU Lille, F-59000, Lille, France
| | - Delphine Ley
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
- Division of Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Jeanne de Flandre Children's Hospital, CHU Lille, F-59000, Lille, France
| | - Jean Lesage
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
| | - Marie Titécat
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
- Laboratory of Bacteriology, Institute of Microbiology, CHU Lille, Lille, France
| |
Collapse
|
10
|
Szyller H, Antosz K, Batko J, Mytych A, Dziedziak M, Wrześniewska M, Braksator J, Pytrus T. Bioactive Components of Human Milk and Their Impact on Child's Health and Development, Literature Review. Nutrients 2024; 16:1487. [PMID: 38794725 PMCID: PMC11124180 DOI: 10.3390/nu16101487] [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: 04/10/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
The composition of human breast milk is an ideal combination of substances necessary for the healthy development of an infant's body while protecting from pathogens and the balanced development of the microbiota. Its composition is dynamic and changes with the age of the child, meeting their current needs. The study provides a thorough overview of human milk components, such as immunological components, growth factors, hormones, carbohydrates, lipids, minerals, and vitamins. Authors focus on capturing the most important aspects of the effects of these substances on a newborn's body, while also looking for specific connections and describing the effects on given systems. Supplementation and the use of ingredients are also discussed. The purpose of this paper is to present the current state of knowledge about the bioactive components of human milk and their impact on the growth, development, and health of the young child.
Collapse
Affiliation(s)
- Hubert Szyller
- Student Scientific Group of Pediatric Gastroenterology and Nutrition, Wroclaw Medical University, 50-369 Wroclaw, Poland; (K.A.); (J.B.); (A.M.); (M.D.); (M.W.)
| | - Katarzyna Antosz
- Student Scientific Group of Pediatric Gastroenterology and Nutrition, Wroclaw Medical University, 50-369 Wroclaw, Poland; (K.A.); (J.B.); (A.M.); (M.D.); (M.W.)
| | - Joanna Batko
- Student Scientific Group of Pediatric Gastroenterology and Nutrition, Wroclaw Medical University, 50-369 Wroclaw, Poland; (K.A.); (J.B.); (A.M.); (M.D.); (M.W.)
| | - Agata Mytych
- Student Scientific Group of Pediatric Gastroenterology and Nutrition, Wroclaw Medical University, 50-369 Wroclaw, Poland; (K.A.); (J.B.); (A.M.); (M.D.); (M.W.)
| | - Marta Dziedziak
- Student Scientific Group of Pediatric Gastroenterology and Nutrition, Wroclaw Medical University, 50-369 Wroclaw, Poland; (K.A.); (J.B.); (A.M.); (M.D.); (M.W.)
| | - Martyna Wrześniewska
- Student Scientific Group of Pediatric Gastroenterology and Nutrition, Wroclaw Medical University, 50-369 Wroclaw, Poland; (K.A.); (J.B.); (A.M.); (M.D.); (M.W.)
| | - Joanna Braksator
- 2nd Clinical Department of Paediatrics, Gastroenterology and Nutrition, Wroclaw Medical University, 50-369 Wrocalw, Poland; (J.B.); (T.P.)
| | - Tomasz Pytrus
- 2nd Clinical Department of Paediatrics, Gastroenterology and Nutrition, Wroclaw Medical University, 50-369 Wrocalw, Poland; (J.B.); (T.P.)
| |
Collapse
|
11
|
Murali SK, Mansell TJ. Next generation probiotics: Engineering live biotherapeutics. Biotechnol Adv 2024; 72:108336. [PMID: 38432422 DOI: 10.1016/j.biotechadv.2024.108336] [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: 09/16/2023] [Revised: 02/10/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
The population dynamics of the human microbiome have been associated with inflammatory bowel disease, cancer, obesity, autoimmune diseases, and many other human disease states. An emerging paradigm in treatment is the administration of live engineered organisms, also called next-generation probiotics. However, the efficacy of these microbial therapies can be limited by the organism's overall performance in the harsh and nutrient-limited environment of the gut. In this review, we summarize the current state of the art use of bacterial and yeast strains as probiotics, highlight the recent development of genetic tools for engineering new therapeutic functions in these organisms, and report on the latest therapeutic applications of engineered probiotics, including recent clinical trials. We also discuss the supplementation of prebiotics as a method of manipulating the microbiome and improving the overall performance of engineered live biotherapeutics.
Collapse
Affiliation(s)
- Sanjeeva Kumar Murali
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA.
| | - Thomas J Mansell
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA; Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
12
|
Mao S, Zhao A, Jiang H, Yan J, Zhong W, Xun Y, Zhang Y. Patterns of Human Milk Oligosaccharides in Mature Milk Are Associated with Certain Gut Microbiota in Infants. Nutrients 2024; 16:1287. [PMID: 38732534 PMCID: PMC11085179 DOI: 10.3390/nu16091287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Human milk oligosaccharides (HMOs) are complexes that play a crucial role in shaping the early-life gut microbiota. This study intends to explore whether HMO patterns are associated with the gut microbiota of infants. We included 96 Chinese breastfeeding mother-infant dyads. Breast milk and infant faecal samples were collected and tested. With milk 2'-fucosyllactose, difucosyllactose, and lacto-N-fucopentaose-I as biomarkers, we divided the mothers into secretor and non-secretor groups. HMO patterns were extracted using principal component analysis. The majority (70.7%) of mothers were categorised as secretor and five different HMO patterns were identified. After adjustment, the infants of secretor mothers exhibited a lower relative abundance of Bifidobacterium bifidum (β = -0.245, 95%CI: -0.465~-0.025). An HMO pattern characterised by high levels of 3-fucosyllactose, lacto-N-fucopentaose-III, and lacto-N-neodifucohexaose-II was positively associated with the relative abundance of Bifidobacterium breve (p = 0.014), while the pattern characterised by lacto-N-neotetraose, 6'-sialyllactose, and sialyllacto-N-tetraose-b was negatively associated with Bifidobacterium breve (p = 0.027). The pattern characterised by high levels of monofucosyl-lacto-N-hexaose-III and monofucosyl-lacto-N-neohexaose was positively associated with Bifidobacterium dentium (p = 0.025) and Bifidobacterium bifidum (p < 0.001), respectively. This study suggests that HMO patterns from mature breast milk were associated with certain gut microbiota of breastfed infants.
Collapse
Affiliation(s)
- Shuai Mao
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (S.M.); (W.Z.)
| | - Ai Zhao
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China;
| | - Hua Jiang
- School of Nursing, Peking University, Beijing 100091, China;
| | - Jingyu Yan
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China;
| | - Wuxian Zhong
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (S.M.); (W.Z.)
| | - Yiping Xun
- Junlebao Dairy Joint Laboratory of Breast Milk Science and Life Health, Peking University, Beijing 100191, China;
| | - Yumei Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (S.M.); (W.Z.)
- Junlebao Dairy Joint Laboratory of Breast Milk Science and Life Health, Peking University, Beijing 100191, China;
| |
Collapse
|
13
|
Gurung M, Schlegel BT, Rajasundaram D, Fox R, Bode L, Yao T, Lindemann SR, LeRoith T, Read QD, Simecka C, Carroll L, Andres A, Yeruva L. Microbiota from human infants consuming secretors or non-secretors mothers' milk impacts the gut and immune system in mice. mSystems 2024; 9:e0029424. [PMID: 38530054 PMCID: PMC11019842 DOI: 10.1128/msystems.00294-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024] Open
Abstract
Maternal secretor status is one of the determinants of human milk oligosaccharides (HMOs) composition, which, in turn, influences the gut microbiota composition of infants. To understand if this change in gut microbiota impacts immune cell composition, intestinal morphology, and gene expression, 21-day-old germ-free C57BL/6 mice were transplanted with fecal microbiota from infants whose mothers were either secretors (SMM) or non-secretors (NSM) or from infants consuming dairy-based formula (MFM). For each group, one set of mice was supplemented with HMOs. HMO supplementation did not significantly impact the microbiota diversity; however, SMM mice had a higher abundance of genus Bacteroides, Bifidobacterium, and Blautia, whereas, in the NSM group, there was a higher abundance of Akkermansia, Enterocloster, and Klebsiella. In MFM, gut microbiota was represented mainly by Parabacteroides, Ruminococcaceae_unclassified, and Clostrodium_sensu_stricto. In mesenteric lymph node, Foxp3+ T cells and innate lymphoid cells type 2 were increased in MFM mice supplemented with HMOs, while in the spleen, they were increased in SMM + HMOs mice. Similarly, serum immunoglobulin A was also elevated in MFM + HMOs group. Distinct global gene expression of the gut was observed in each microbiota group, which was enhanced with HMOs supplementation. Overall, our data show that distinct infant gut microbiota due to maternal secretor status or consumption of dairy-based formula and HMO supplementation impacts immune cell composition, antibody response, and intestinal gene expression in a mouse model. IMPORTANCE Early life factors like neonatal diet modulate gut microbiota, which is important for the optimal gut and immune function. One such factor, human milk oligosaccharides (HMOs), the composition of which is determined by maternal secretor status, has a profound effect on infant gut microbiota. However, how the infant gut microbiota composition determined by maternal secretor status or consumption of infant formula devoid of HMOs impacts infant intestinal ammorphology, gene expression, and immune signature is not well explored. This study provides insights into the differential establishment of infant microbiota derived from infants fed by secretor or non-secretor mothers milk or those consuming infant formula and demonstrates that the secretor status of mothers promotes Bifidobacteria and Bacteroides sps. establishment. This study also shows that supplementation of pooled HMOs in mice changed immune cell composition in the spleen and mesenteric lymph nodes and immunoglobulins in circulation. Hence, this study highlights that maternal secretor status has a role in infant gut microbiota composition, and this, in turn, can impact host gut and immune system.
Collapse
Affiliation(s)
- Manoj Gurung
- Microbiome and Metabolism Research Unit (MMRU), USDA-ARS, SEA, Arkansas Children’s Nutrition Center, Little Rock, Arkansas, USA
| | - Brent Thomas Schlegel
- University of Pittsburgh Medical Center (UPMC), Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Dhivyaa Rajasundaram
- University of Pittsburgh Medical Center (UPMC), Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Renee Fox
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Lars Bode
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, California, USA
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Tianming Yao
- Department of Food Science, Whistler Center for Carbohydrate Research, Purdue University, West Lafayette, Indiana, USA
| | - Stephen R. Lindemann
- Department of Food Science, Whistler Center for Carbohydrate Research, Purdue University, West Lafayette, Indiana, USA
| | - Tanya LeRoith
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, Virginia, USA
| | | | - Christy Simecka
- Division of Laboratory Animal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Laura Carroll
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Aline Andres
- Microbiome and Metabolism Research Unit (MMRU), USDA-ARS, SEA, Arkansas Children’s Nutrition Center, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Laxmi Yeruva
- Microbiome and Metabolism Research Unit (MMRU), USDA-ARS, SEA, Arkansas Children’s Nutrition Center, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
14
|
Lordan C, Roche AK, Delsing D, Nauta A, Groeneveld A, MacSharry J, Cotter PD, van Sinderen D. Linking human milk oligosaccharide metabolism and early life gut microbiota: bifidobacteria and beyond. Microbiol Mol Biol Rev 2024; 88:e0009423. [PMID: 38206006 PMCID: PMC10966949 DOI: 10.1128/mmbr.00094-23] [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: 01/12/2024] Open
Abstract
SUMMARYHuman milk oligosaccharides (HMOs) are complex, multi-functional glycans present in human breast milk. They represent an intricate mix of heterogeneous structures which reach the infant intestine in an intact form as they resist gastrointestinal digestion. Therefore, they confer a multitude of benefits, directly and/or indirectly, to the developing neonate. Certain bifidobacterial species, being among the earliest gut colonizers of breast-fed infants, have an adapted functional capacity to metabolize various HMO structures. This ability is typically observed in infant-associated bifidobacteria, as opposed to bifidobacteria associated with a mature microbiota. In recent years, information has been gleaned regarding how these infant-associated bifidobacteria as well as certain other taxa are able to assimilate HMOs, including the mechanistic strategies enabling their acquisition and consumption. Additionally, complex metabolic interactions occur between microbes facilitated by HMOs, including the utilization of breakdown products released from HMO degradation. Interest in HMO-mediated changes in microbial composition and function has been the focal point of numerous studies, in recent times fueled by the availability of individual biosynthetic HMOs, some of which are now commonly included in infant formula. In this review, we outline the main HMO assimilatory and catabolic strategies employed by infant-associated bifidobacteria, discuss other taxa that exhibit breast milk glycan degradation capacity, and cover HMO-supported cross-feeding interactions and related metabolites that have been described thus far.
Collapse
Affiliation(s)
- Cathy Lordan
- Teagasc Food Research Centre, Fermoy, Co Cork, Ireland
| | - Aoife K. Roche
- APC Microbiome Ireland, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | | | - Arjen Nauta
- FrieslandCampina, Amersfoort, the Netherlands
| | | | - John MacSharry
- APC Microbiome Ireland, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Paul D. Cotter
- Teagasc Food Research Centre, Fermoy, Co Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Douwe van Sinderen
- APC Microbiome Ireland, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
15
|
Belei O, Basaca DG, Olariu L, Pantea M, Bozgan D, Nanu A, Sîrbu I, Mărginean O, Enătescu I. The Interaction between Stress and Inflammatory Bowel Disease in Pediatric and Adult Patients. J Clin Med 2024; 13:1361. [PMID: 38592680 PMCID: PMC10932475 DOI: 10.3390/jcm13051361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 04/10/2024] Open
Abstract
Background: Inflammatory bowel diseases (IBDs) have seen an exponential increase in incidence, particularly among pediatric patients. Psychological stress is a significant risk factor influencing the disease course. This review assesses the interaction between stress and disease progression, focusing on articles that quantified inflammatory markers in IBD patients exposed to varying degrees of psychological stress. Methods: A systematic narrative literature review was conducted, focusing on the interaction between IBD and stress among adult and pediatric patients, as well as animal subjects. The research involved searching PubMed, Scopus, Medline, and Cochrane Library databases from 2000 to December 2023. Results: The interplay between the intestinal immunity response, the nervous system, and psychological disorders, known as the gut-brain axis, plays a major role in IBD pathophysiology. Various types of stressors alter gut mucosal integrity through different pathways, increasing gut mucosa permeability and promoting bacterial translocation. A denser microbial load in the gut wall emphasizes cytokine production, worsening the disease course. The risk of developing depression and anxiety is higher in IBD patients compared with the general population, and stress is a significant trigger for inducing acute flares of the disease. Conclusions: Further large studies should be conducted to assess the relationship between stressors, psychological disorders, and their impact on the course of IBD. Clinicians involved in the medical care of IBD patients should aim to implement stress reduction practices in addition to pharmacological therapies.
Collapse
Affiliation(s)
- Oana Belei
- First Pediatric Clinic, Disturbances of Growth and Development on Children Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (O.B.); (O.M.)
- Department of Pediatrics, First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Diana-Georgiana Basaca
- First Pediatric Clinic, Disturbances of Growth and Development on Children Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (O.B.); (O.M.)
- Department of Pediatrics, First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Laura Olariu
- Department of Pediatrics, First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Manuela Pantea
- Twelfth Department, Neonatology Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (M.P.); (I.E.)
| | - Daiana Bozgan
- Clinic of Neonatology, “Pius Brânzeu” County Emergency Clinical Hospital, 300723 Timișoara, Romania;
| | - Anda Nanu
- Third Pediatric Clinic, “Louis Țurcanu” Emergency Children Hospital, 300011 Timișoara, Romania; (A.N.); (I.S.)
| | - Iuliana Sîrbu
- Third Pediatric Clinic, “Louis Țurcanu” Emergency Children Hospital, 300011 Timișoara, Romania; (A.N.); (I.S.)
| | - Otilia Mărginean
- First Pediatric Clinic, Disturbances of Growth and Development on Children Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (O.B.); (O.M.)
- Department of Pediatrics, First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Ileana Enătescu
- Twelfth Department, Neonatology Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (M.P.); (I.E.)
| |
Collapse
|
16
|
Núñez-Delgado A, Mizrachi-Chávez VM, Welti-Chanes J, Macher-Quintana ST, Chuck-Hernández C. Breast milk preservation: thermal and non-thermal processes and their effect on microorganism inactivation and the content of bioactive and nutritional compounds. Front Nutr 2024; 10:1325863. [PMID: 38455872 PMCID: PMC10919153 DOI: 10.3389/fnut.2023.1325863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 12/26/2023] [Indexed: 03/09/2024] Open
Abstract
Human Breast Milk (HBM) is widely acknowledged as the best nutritional source for neonates. Data indicates that, in 2019, 83.2% of infants in the United States received breast milk at birth, slightly reducing to 78.6% at 1 month. Despite these encouraging early figures, exclusive breastfeeding rates sharply declined, dropping to 24.9% by 6 months. This decline is particularly pronounced when direct breastfeeding is challenging, such as in Neonatal Intensive Care Units (NICU) and for working mothers. Given this, it is vital to explore alternative breast milk preservation methods. Technologies like Holder Pasteurization (HoP), High-Temperature Short-Time Pasteurization (HTST), High-Pressure Processing (HPP), UV radiation (UV), and Electric Pulses (PEF) have been introduced to conserve HBM. This review aims to enhance the understanding of preservation techniques for HBM, supporting the practice of extended exclusive breastfeeding. It explicitly addresses microbial concerns, focusing on critical pathogens like Staphylococcus aureus, Enterococcus, Escherichia coli, Listeria monocytogenes, and Cytomegalovirus, and explores how various preservation methods can mitigate these risks. Additionally, the review highlights the importance of retaining the functional elements of HBM, particularly its immunological components such as antibodies and enzymes like lysozyme and Bile Salt Stimulated Lipase (BSSL). The goal is to provide a comprehensive overview of the current state of HBM treatment, critically assess existing practices, identify areas needing improvement, and advocate for extended exclusive breastfeeding due to its vital role in ensuring optimal nutrition and overall health in infants.
Collapse
Affiliation(s)
- Alejandro Núñez-Delgado
- Tecnologico, de Monterrey, School of Engineering and Sciences, Monterrey, Mexico
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, Mexico
| | | | - Jorge Welti-Chanes
- Tecnologico, de Monterrey, School of Engineering and Sciences, Monterrey, Mexico
| | | | | |
Collapse
|
17
|
Le Bourgot C, Lollier V, Richer Y, Thoulouze L, Svilar L, Le Gall S, Blat S, Le Huërou-Luron I. Maternal short chain fructo-oligosaccharides supplementation during late gestation and lactation influences milk components and offspring gut metabolome: a pilot study. Sci Rep 2024; 14:4236. [PMID: 38378944 PMCID: PMC10879084 DOI: 10.1038/s41598-024-54813-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 02/16/2024] [Indexed: 02/22/2024] Open
Abstract
Breast milk composition is influenced by maternal diet. This study aimed to evaluate if supplementation of maternal diet with a prebiotic fibre, through its potential effect on milk composition, can be a leverage to orientate the gut microbiota of infants in a way that would be beneficial for their health. Twelve sows received a diet supplemented with short chain fructo-oligosaccharides or maltodextrins during the last month of gestation and the lactation. Oligosaccharidic and lipidomic profiles of colostrum and mature milk (21 days), as well as faecal microbiota composition and metabolomic profile of 21 day-old piglets were evaluated. The total porcine milk oligosaccharide concentration tended to be lower in scFOS-supplemented sows, mainly due to the significant reduction of the neutral core oligosaccharides (in particular that of a tetrahexose). Maternal scFOS supplementation affected the concentration of 31 lipids (mainly long-chain triglycerides) in mature milk. Faecal short-chain fatty acid content and that of 16 bacterial metabolites were modified by scFOS supplementation. Interestingly, the integrative data analysis gave a novel insight into the relationships between (i) maternal milk lipids and PMOs and (ii) offspring faecal bacteria and metabolites. In conclusion, scFOS-enriched maternal diet affected the composition of mature milk, and this was associated with a change in the colonisation of the offspring intestinal microbiota.
Collapse
Affiliation(s)
- Cindy Le Bourgot
- Tereos, Scientific and Regulatory Affairs Department, Moussy-le-Vieux, France.
| | - Virginie Lollier
- INRAE, UR1268 BIA, 44300, Nantes, France
- INRAE, PROBE Research Infrastructure, BIBS Facility, 44300, Nantes, France
| | - Yoann Richer
- INRAE, UR1268 BIA, 44300, Nantes, France
- INRAE, PROBE Research Infrastructure, BIBS Facility, 44300, Nantes, France
| | - Loric Thoulouze
- INRAE, UR1268 BIA, 44300, Nantes, France
- INRAE, PROBE Research Infrastructure, BIBS Facility, 44300, Nantes, France
| | - Ljubica Svilar
- Cribiom, Centre de Recherche Cardiovasculaire et Nutrition C2VN, UMR INRAE 1260 INSERM 1263, University Aix-Marseille, Marseille, France
| | - Sophie Le Gall
- INRAE, UR1268 BIA, 44300, Nantes, France
- INRAE, PROBE Research Infrastructure, BIBS Facility, 44300, Nantes, France
| | - Sophie Blat
- Institut NuMeCan, INRAE, INSERM, University Rennes, 35590, Saint-Gilles, France
| | | |
Collapse
|
18
|
Wang X, Li L, Liu T, Shi Y. More than nutrition: Therapeutic potential and mechanism of human milk oligosaccharides against necrotizing enterocolitis. Life Sci 2024; 339:122420. [PMID: 38218534 DOI: 10.1016/j.lfs.2024.122420] [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: 10/22/2023] [Revised: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 01/15/2024]
Abstract
Human milk is the most valuable source of nutrition for infants. The structure and function of human milk oligosaccharides (HMOs), which are key components of human milk, have long been attracting particular research interest. Several recent studies have found HMOs to be efficacious in the prevention and treatment of necrotizing enterocolitis (NEC). Additionally, they could be developed in the future as non-invasive predictive markers for NEC. Based on previous findings and the well-defined functions of HMOs, we summarize potential protective mechanisms of HMOs against neonatal NEC, which include: modulating signal receptor function, promoting intestinal epithelial cell proliferation, reducing apoptosis, restoring intestinal blood perfusion, regulating microbial prosperity, and alleviating intestinal inflammation. HMOs supplementation has been demonstrated to be protective against NEC in both animal studies and clinical observations. This calls for mass production and use of HMOs in infant formula, necessitating more research into the safety of industrially produced HMOs and the appropriate dosage in infant formula.
Collapse
Affiliation(s)
- Xinru Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, San Hao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Ling Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, San Hao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Tianjing Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, San Hao Street, Heping District, Shenyang, Liaoning 110004, China.
| | - Yongyan Shi
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, San Hao Street, Heping District, Shenyang, Liaoning 110004, China.
| |
Collapse
|
19
|
Aljahdali AA, Cantoral A, Peterson KE, Perng W, Mercado-García A, Téllez-Rojo MM, Ramírez-Silva CI, Jansen EC. Breastfeeding Duration and Cardiometabolic Health during Adolescence: A Longitudinal Analysis. J Pediatr 2024; 265:113768. [PMID: 37802388 DOI: 10.1016/j.jpeds.2023.113768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/04/2023] [Accepted: 09/29/2023] [Indexed: 10/10/2023]
Abstract
OBJECTIVE To investigate the longitudinal association between breastfeeding duration and cardiometabolic health, using repeated measures study design among children and adolescents. STUDY DESIGN This study included 634 offsprings aged 10 to 21 years (52% female) from the Early Life Exposure in Mexico to Environmental Toxicants birth cohort followed up to four time points during adolescence. Breastfeeding duration was prospectively quantified using questionnaires during early childhood. Cardiometabolic risk factors, body composition, and weight-related biomarkers were assessed as outcomes during adolescent follow-up visits. Sex-stratified linear mixed-effects models were used to model the association between quartiles of breastfeeding duration and outcomes, adjusting for age and additional covariates. RESULTS Median breastfeeding duration was 7 months (minimum = 0, maximum = 36). Boys in the second quartile (median breastfeeding = 5 months) had lower total fat mass % (β (SE) -3.2 (1.5) P = .037), and higher lean mass % (3.1 (1.6) P = .049) and skeletal muscle mass % (1.8 (0.8) P = .031) compared with the reference group (median breastfeeding = 2 months). A positive linear trend between breastfeeding duration and trunk lean mass % (0.1 (0.04) P = .035) was found among girls. No association was found with other cardiometabolic indicators. CONCLUSION Despite sex-specific associations of breastfeeding duration with body composition, there was a lack of substantial evidence for the protective effects of breastfeeding against impaired cardiometabolic health during adolescence among Mexican youth. Further longitudinal studies with a robust assessment of breastfeeding are recommended.
Collapse
Affiliation(s)
- Abeer A Aljahdali
- Department of Clinical Nutrition, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI
| | | | - Karen E Peterson
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI; Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI.
| | - Wei Perng
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Adriana Mercado-García
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Mexico
| | - Martha M Téllez-Rojo
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Mexico
| | | | - Erica C Jansen
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI
| |
Collapse
|
20
|
Capeding MRZ, Phee LCM, Ming C, Noti M, Vidal K, Le Carrou G, Frézal A, Moll JM, Vogt JK, Myers PN, Nielsen BH, Boulangé CL, Samuel TM, Berger B, Cercamondi CI. Safety, efficacy, and impact on gut microbial ecology of a Bifidobacterium longum subspecies infantis LMG11588 supplementation in healthy term infants: a randomized, double-blind, controlled trial in the Philippines. Front Nutr 2023; 10:1319873. [PMID: 38162520 PMCID: PMC10755859 DOI: 10.3389/fnut.2023.1319873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/16/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction Bifidobacterium longum subspecies infantis (B. infantis) may play a key role in infant gut development. This trial evaluated safety, tolerability, and efficacy of B. infantis LMG11588 supplementation. Methods This randomized, placebo-controlled, double-blind study conducted in the Philippines included healthy breastfed and/or formula-fed infants (14-21 days old) randomized for 8 weeks to a control group (CG; n = 77), or any of two B. infantis experimental groups (EGs): low (Lo-EG; 1*108 CFU/day; n = 75) or high dose (Hi-EG; 1.8*1010 CFU/day; n = 76). Primary endpoint was weight gain; secondary endpoints included stooling patterns, gastrointestinal symptoms, adverse events, fecal microbiome, biomarkers, pH, and organic acids. Results Non-inferiority in weight gain was demonstrated for Hi-EG and Lo-EG vs. CG. Overall, probiotic supplementation promoted mushy-soft stools, fewer regurgitation episodes, and increased fecal acetate production, which was more pronounced in the exclusively breastfed infants (EBF) and positively correlated with B. infantis abundance. In EBF, fecal pro-inflammatory cytokines (IL-1 beta, IL-8) were reduced. Strain-level metagenomic analysis allowed attributing the increased abundance of B. infantis in EGs versus CG, to LMG11588 probiotic colonization. Colonization by autochthonous B. infantis strains was similar between groups. Discussion B. infantis LMG11588 supplementation was associated with normal infant growth, was safe and well-tolerated and promoted a Bifidobacterium-rich microbiota driven by B. infantis LMG11588 colonization without disturbing the natural dispersal of autochthonous B. infantis strains. In EBF, supplementation stimulated microbial metabolic activity and beneficially modulated enteric inflammation.
Collapse
Affiliation(s)
| | | | - Chang Ming
- Biostatistics & Data, Nestlé Research, Lausanne, Switzerland
| | - Mario Noti
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Karine Vidal
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Gilles Le Carrou
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - A. Frézal
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | | | | | | | | | - Claire L. Boulangé
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Tinu Mary Samuel
- Nestlé Product Technology Center – Nutrition, Société des Produits Nestlé S.A., Vevey, Switzerland
| | - Bernard Berger
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Colin Ivano Cercamondi
- Nestlé Product Technology Center – Nutrition, Société des Produits Nestlé S.A., Vevey, Switzerland
| |
Collapse
|
21
|
Dubernat L, Marousez L, Desseyn JL, Gouyer V, Hermann E, Gottrand F, Ley D, Lesage J. [Human milk oligosaccharides play major roles in child development and future health]. Med Sci (Paris) 2023; 39:869-875. [PMID: 38018931 DOI: 10.1051/medsci/2023164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023] Open
Abstract
Human milk oligosaccharides (HMO) represent the third largest component of human breast milk (BM). The BM level is comprised between 5 to 20 g per liter and they have a great structural complexity with more than 150 HMO characterized to date. In this review, we present a summary of the main experimental and clinical data that have demonstrated their multiple biological roles in infants such as for gut development, microbiota, immune protection and neurodevelopment. Some HMO-enriched infant formulas are available yet, even if their benefits on the infant health remain to be confirmed. Further researches could allow therapeutic use in preterm newborns or in infants with intestinal diseases. Experimental data suggest that they could also be used in the prevention of some chronic diseases with immunometabolic or neurodevelopmental components.
Collapse
Affiliation(s)
- Laure Dubernat
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE, F-59000 Lille, France
| | - Lucie Marousez
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE, F-59000 Lille, France
| | - Jean-Luc Desseyn
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE, F-59000 Lille, France
| | - Valérie Gouyer
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE, F-59000 Lille, France
| | - Emmanuel Hermann
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE, F-59000 Lille, France
| | - Frédéric Gottrand
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE, F-59000 Lille, France - Service de gastroentérologie, hépatologie et nutrition, département de pédiatrie, hôpital Jeanne de Flandre, CHU Lille, F-59000 Lille, France
| | - Delphine Ley
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE, F-59000 Lille, France - Service de gastroentérologie, hépatologie et nutrition, département de pédiatrie, hôpital Jeanne de Flandre, CHU Lille, F-59000 Lille, France
| | - Jean Lesage
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE, F-59000 Lille, France
| |
Collapse
|
22
|
Nogacka AM, Cuesta I, Gueimonde M, de los Reyes-Gavilán CG. 2-Fucosyllactose Metabolism by Bifidobacteria Promotes Lactobacilli Growth in Co-Culture. Microorganisms 2023; 11:2659. [PMID: 38004671 PMCID: PMC10673426 DOI: 10.3390/microorganisms11112659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Breastfeeding is recognized as the gold standard in infant nutrition, not only because of breastmilk's intrinsic nutritional benefits but also due to the high content of different bioactive components such as 2-fucosyllactose (2'FL) in the mother's milk. It promotes the growth of its two major consumers, Bifidobacterium longum ssp. infantis and Bifidobacterium bifidum, but the effect on other intestinal microorganisms of infant microbiota remains incompletely understood. pH-uncontrolled fecal cultures from infants donors identified as "fast 2'FL -degrader" microbiota phenotype were used for the isolation of 2'FL-associated microorganisms. The use of specific selective agents allowed the successful isolation of B. bifidum IPLA20048 and of Lactobacillus gasseri IPLA20136. The characterization of 2'FL consumption and its moieties has revealed more pronounced growth, pH drop, and lactic acid production after 2'FL consumption when both microorganisms were grown together. The results point to an association between B. bifidum IPLA20048 and L. gasseri IPLA20136 in which L. gasseri is able to use the galactose from the lactose moiety after the hydrolysis of 2'FL by B. bifidum. The additional screening of two groups of bifidobacteria (n = 38), fast and slow degraders of 2'FL, in co-culture with lactobacilli confirmed a potential cross-feeding mechanism based on degradation products released from bifidobacterial 2'FL break-down. Our work suggests that this phenomenon may be widespread among lactobacilli and bifidobacteria in the infant gut. More investigation is needed to decipher how the ability to degrade 2'FL and other human milk oligosaccharides could influence the microbiota establishment in neonates and the evolution of the microbiota in adult life.
Collapse
Affiliation(s)
- Alicja M. Nogacka
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (I.C.); (M.G.); (C.G.d.l.R.-G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| | - Isabel Cuesta
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (I.C.); (M.G.); (C.G.d.l.R.-G.)
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (I.C.); (M.G.); (C.G.d.l.R.-G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (I.C.); (M.G.); (C.G.d.l.R.-G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| |
Collapse
|
23
|
Hackenbruch SN, Cutajar D, Farrugia RD. Enteral nutrition of preterm infants on the Maltese islands: are we doing it right? Eur J Clin Nutr 2023; 77:1005-1007. [PMID: 37419970 DOI: 10.1038/s41430-023-01304-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 05/23/2023] [Accepted: 06/20/2023] [Indexed: 07/09/2023]
Affiliation(s)
| | - Daniel Cutajar
- Department of Medicine, Mater Die Hospital, MSD2090, Msida, Malta
| | - Ryan Dominic Farrugia
- Department of Child and Adolescent Health, Mater Die Hospital, MSD2090, Msida, Malta
| |
Collapse
|
24
|
Lis-Kuberka J, Pupek M, Orczyk-Pawiłowicz M. The Mother-Child Dyad Adipokine Pattern: A Review of Current Knowledge. Nutrients 2023; 15:4059. [PMID: 37764842 PMCID: PMC10535905 DOI: 10.3390/nu15184059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
An important role in the network of interconnections between the mother and child is played by adipokines, which are adipose tissue hormones engaged in the regulation of metabolism. Alternations of maternal adipokines translate to the worsening of maternal insulin resistance as well as metabolic stress, altered placenta functions, and fetal development, which finally contribute to long-term metabolic unfavorable conditions. This paper is the first to summarize the current state of knowledge concerning the concentrations of individual adipokines in different biological fluids of maternal and cord plasma, newborn/infant plasma, milk, and the placenta, where it highlights the impact of adverse perinatal risk factors, including gestational diabetes mellitus, preeclampsia, intrauterine growth restriction, preterm delivery, and maternal obesity on the adipokine patterns in maternal-infant dyads. The importance of adipokine measurement and relationships in biological fluids during pregnancy and lactation is crucial for public health in the area of prevention of most diet-related metabolic diseases. The review highlights the huge knowledge gap in the field of hormones participating in the energy homeostasis and metabolic pathways during perinatal and postnatal periods in the mother-child dyad. An in-depth characterization is needed to confirm if the adverse outcomes of early developmental programming might be modulated via maternal lifestyle intervention.
Collapse
Affiliation(s)
- Jolanta Lis-Kuberka
- Department of Biochemistry and Immunochemistry, Division of Chemistry and Immunochemistry, Wroclaw Medical University, M. Skłodowskiej-Curie 48/50, 50-369 Wroclaw, Poland
| | | | - Magdalena Orczyk-Pawiłowicz
- Department of Biochemistry and Immunochemistry, Division of Chemistry and Immunochemistry, Wroclaw Medical University, M. Skłodowskiej-Curie 48/50, 50-369 Wroclaw, Poland
| |
Collapse
|
25
|
Bastos-Moreira Y, Ouédraogo L, De Boevre M, Argaw A, de Kok B, Hanley-Cook GT, Deng L, Ouédraogo M, Compaoré A, Tesfamariam K, Ganaba R, Huybregts L, Toe LC, Lachat C, Kolsteren P, De Saeger S, Dailey-Chwalibóg T. A Multi-Omics and Human Biomonitoring Approach to Assessing the Effectiveness of Fortified Balanced Energy-Protein Supplementation on Maternal and Newborn Health in Burkina Faso: A Study Protocol. Nutrients 2023; 15:4056. [PMID: 37764838 PMCID: PMC10535470 DOI: 10.3390/nu15184056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Fortified balanced energy-protein (BEP) supplementation is a promising intervention for improving maternal health, birth outcomes and infant growth in low- and middle-income countries. This nested biospecimen sub-study aimed to evaluate the physiological effect of multi-micronutrient-fortified BEP supplementation on pregnant and lactating women and their infants. Pregnant women (15-40 years) received either fortified BEP and iron-folic acid (IFA) (intervention) or IFA only (control) throughout pregnancy. The same women were concurrently randomized to receive either a fortified BEP supplement during the first 6 months postpartum in combination with IFA for the first 6 weeks (i.e., intervention) or the postnatal standard of care, which comprised IFA alone for 6 weeks postpartum (i.e., control). Biological specimens were collected at different timepoints. Multi-omics profiles will be characterized to assess the mediating effect of BEP supplementation on the different trial arms and its effect on maternal health, as well as birth and infant growth outcomes. The mediating effect of the exposome in the relationship between BEP supplementation and maternal health, birth outcomes and infant growth were characterized via biomonitoring markers of air pollution, mycotoxins and environmental contaminants. The results will provide holistic insight into the granular physiological effects of prenatal and postnatal BEP supplementation.
Collapse
Affiliation(s)
- Yuri Bastos-Moreira
- Center of Excellence in Mycotoxicology and Public Health, MYTOXSOUTH Coordination Unit, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; (M.D.B.); (S.D.S.)
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.); (A.A.); (B.d.K.); (G.T.H.-C.); (L.D.); (K.T.); (L.H.); (L.C.T.); (C.L.); (P.K.)
| | - Lionel Ouédraogo
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.); (A.A.); (B.d.K.); (G.T.H.-C.); (L.D.); (K.T.); (L.H.); (L.C.T.); (C.L.); (P.K.)
- Centre Muraz, Bobo-Dioulasso 01 BP 390, Burkina Faso
| | - Marthe De Boevre
- Center of Excellence in Mycotoxicology and Public Health, MYTOXSOUTH Coordination Unit, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; (M.D.B.); (S.D.S.)
| | - Alemayehu Argaw
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.); (A.A.); (B.d.K.); (G.T.H.-C.); (L.D.); (K.T.); (L.H.); (L.C.T.); (C.L.); (P.K.)
| | - Brenda de Kok
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.); (A.A.); (B.d.K.); (G.T.H.-C.); (L.D.); (K.T.); (L.H.); (L.C.T.); (C.L.); (P.K.)
| | - Giles T. Hanley-Cook
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.); (A.A.); (B.d.K.); (G.T.H.-C.); (L.D.); (K.T.); (L.H.); (L.C.T.); (C.L.); (P.K.)
| | - Lishi Deng
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.); (A.A.); (B.d.K.); (G.T.H.-C.); (L.D.); (K.T.); (L.H.); (L.C.T.); (C.L.); (P.K.)
| | - Moctar Ouédraogo
- Agence de Formation de Recherche et d’Expertise en Santé pour l’Afrique (AFRICSanté), Bobo-Dioulasso 01 BP 298, Burkina Faso; (M.O.); (A.C.); (R.G.)
| | - Anderson Compaoré
- Agence de Formation de Recherche et d’Expertise en Santé pour l’Afrique (AFRICSanté), Bobo-Dioulasso 01 BP 298, Burkina Faso; (M.O.); (A.C.); (R.G.)
| | - Kokeb Tesfamariam
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.); (A.A.); (B.d.K.); (G.T.H.-C.); (L.D.); (K.T.); (L.H.); (L.C.T.); (C.L.); (P.K.)
| | - Rasmané Ganaba
- Agence de Formation de Recherche et d’Expertise en Santé pour l’Afrique (AFRICSanté), Bobo-Dioulasso 01 BP 298, Burkina Faso; (M.O.); (A.C.); (R.G.)
| | - Lieven Huybregts
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.); (A.A.); (B.d.K.); (G.T.H.-C.); (L.D.); (K.T.); (L.H.); (L.C.T.); (C.L.); (P.K.)
- Nutrition, Diets, and Health Unit, Department of Food and Nutrition Policy, International Food Policy Research Institute (IFPRI), Washington, DC 20005, USA
| | - Laeticia Celine Toe
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.); (A.A.); (B.d.K.); (G.T.H.-C.); (L.D.); (K.T.); (L.H.); (L.C.T.); (C.L.); (P.K.)
- Unité Nutrition et Maladies Métaboliques, Institut de Recherche en Sciences de la Santé (IRSS), Bobo-Dioulasso 01 BP 545, Burkina Faso
| | - Carl Lachat
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.); (A.A.); (B.d.K.); (G.T.H.-C.); (L.D.); (K.T.); (L.H.); (L.C.T.); (C.L.); (P.K.)
| | - Patrick Kolsteren
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.); (A.A.); (B.d.K.); (G.T.H.-C.); (L.D.); (K.T.); (L.H.); (L.C.T.); (C.L.); (P.K.)
| | - Sarah De Saeger
- Center of Excellence in Mycotoxicology and Public Health, MYTOXSOUTH Coordination Unit, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; (M.D.B.); (S.D.S.)
- Department of Biotechnology and Food Technology, Faculty of Science, University of Johannesburg, Doornfontein Campus, Gauteng 2028, South Africa
| | - Trenton Dailey-Chwalibóg
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (L.O.); (A.A.); (B.d.K.); (G.T.H.-C.); (L.D.); (K.T.); (L.H.); (L.C.T.); (C.L.); (P.K.)
| |
Collapse
|
26
|
Arishi RA, Lai CT, Geddes DT, Stinson LF. Impact of breastfeeding and other early-life factors on the development of the oral microbiome. Front Microbiol 2023; 14:1236601. [PMID: 37744908 PMCID: PMC10513450 DOI: 10.3389/fmicb.2023.1236601] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/25/2023] [Indexed: 09/26/2023] Open
Abstract
The oral cavity is home to the second most diverse microbiome in the human body. This community contributes to both oral and systemic health. Acquisition and development of the oral microbiome is a dynamic process that occurs over early life; however, data regarding longitudinal assembly of the infant oral microbiome is scarce. While numerous factors have been associated with the composition of the infant oral microbiome, early feeding practices (breastfeeding and the introduction of solids) appear to be the strongest determinants of the infant oral microbiome. In the present review, we draw together data on the maternal, infant, and environmental factors linked to the composition of the infant oral microbiome, with a focus on early nutrition. Given evidence that breastfeeding powerfully shapes the infant oral microbiome, the review explores potential mechanisms through which human milk components, including microbes, metabolites, oligosaccharides, and antimicrobial proteins, may interact with and shape the infant oral microbiome. Infancy is a unique period for the oral microbiome. By enhancing our understanding of oral microbiome assembly in early life, we may better support both oral and systemic health throughout the lifespan.
Collapse
Affiliation(s)
- Roaa A. Arishi
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
- Ministry of Health, Riyadh, Saudi Arabia
| | - Ching T. Lai
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
| | - Donna T. Geddes
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
| | - Lisa F. Stinson
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
27
|
Jin C, Lundstrøm J, Korhonen E, Luis AS, Bojar D. Breast Milk Oligosaccharides Contain Immunomodulatory Glucuronic Acid and LacdiNAc. Mol Cell Proteomics 2023; 22:100635. [PMID: 37597722 PMCID: PMC10509713 DOI: 10.1016/j.mcpro.2023.100635] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/31/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023] Open
Abstract
Breast milk is abundant with functionalized milk oligosaccharides (MOs) to nourish and protect the neonate. Yet we lack a comprehensive understanding of the repertoire and evolution of MOs across Mammalia. We report ∼400 MO-species associations (>100 novel structures) from milk glycomics of nine mostly understudied species: alpaca, beluga whale, black rhinoceros, bottlenose dolphin, impala, L'Hoest's monkey, pygmy hippopotamus, domestic sheep, and striped dolphin. This revealed the hitherto unknown existence of the LacdiNAc motif (GalNAcβ1-4GlcNAc) in MOs of all species except alpaca, sheep, and striped dolphin, indicating the widespread occurrence of this potentially antimicrobial motif in MOs. We also characterize glucuronic acid-containing MOs in the milk of impala, dolphins, sheep, and rhinoceros, previously only reported in cows. We demonstrate that these GlcA-MOs exhibit potent immunomodulatory effects. Our study extends the number of known MOs by >15%. Combined with >1900 curated MO-species associations, we characterize MO motif distributions, presenting an exhaustive overview of MO biodiversity.
Collapse
Affiliation(s)
- Chunsheng Jin
- Proteomics Core Facility at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jon Lundstrøm
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Emma Korhonen
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Ana S Luis
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Daniel Bojar
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
28
|
Zhao K, Pang H, Shao K, Yang Z, Li S, He N. The function of human milk oligosaccharides and their substitute oligosaccharides as probiotics in gut inflammation. Food Funct 2023; 14:7780-7798. [PMID: 37575049 DOI: 10.1039/d3fo02092d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Gut inflammation seriously affects the healthy life of patients, and has a trend of increasing incidence rate. However, the current methods for treating gut inflammation are limited to surgery and drugs, which can cause irreversible damage to patients, especially infants. As natural oligosaccharides in human breast milk, human milk oligosaccharides (HMOs) function as probiotics in treating and preventing gut inflammation: improving the abundance of the gut microbiota, increasing the gut barrier function, and reducing the gut inflammatory reaction. Meanwhile, due to the complexity and high cost of their synthesis, people are searching for functional oligosaccharides that can replace HMOs as a food additive in infants milk powder and adjuvant therapy for chronic inflammation. The purpose of this review is to summarize the therapeutic and preventive effects of HMOs and their substitute functional oligosaccharides as probiotics in gut inflammation, and to summarize the prospect of their application in infant breast milk replacement in the future.
Collapse
Affiliation(s)
- Kunyi Zhao
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266003, China.
| | - Hao Pang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266003, China.
| | - Kaidi Shao
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266003, China.
| | - Zizhen Yang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266003, China.
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266003, China.
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266003, China.
| |
Collapse
|
29
|
Schönknecht YB, Moreno Tovar MV, Jensen SR, Parschat K. Clinical Studies on the Supplementation of Manufactured Human Milk Oligosaccharides: A Systematic Review. Nutrients 2023; 15:3622. [PMID: 37630811 PMCID: PMC10458772 DOI: 10.3390/nu15163622] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/12/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Human milk oligosaccharides (HMOs) are a major component of human milk. They are associated with multiple health benefits and are manufactured on a large scale for their addition to different food products. In this systematic review, we evaluate the health outcomes of published clinical trials involving the supplementation of manufactured HMOs. We screened the PubMed database and Cochrane Library, identifying 26 relevant clinical trials and five publications describing follow-up studies. The clinical trials varied in study populations, including healthy term infants, infants with medical indications, children, and adults. They tested eight different HMO structures individually or as blends in varying doses. All trials included safety and tolerance assessments, and some also assessed growth, stool characteristics, infections, gut microbiome composition, microbial metabolites, and biomarkers. The studies consistently found that HMO supplementation was safe and well tolerated. Infant studies reported a shift in outcomes towards those observed in breastfed infants, including stool characteristics, gut microbiome composition, and intestinal immune markers. Beneficial gut health and immune system effects have also been observed in other populations following HMO supplementation. Further clinical trials are needed to substantiate the effects of HMO supplementation on human health and to understand their structure and dose dependency.
Collapse
|
30
|
Bai GH, Tsai MC, Lin SC, Hsu YH, Chen SY. Unraveling the interplay between norovirus infection, gut microbiota, and novel antiviral approaches: a comprehensive review. Front Microbiol 2023; 14:1212582. [PMID: 37485533 PMCID: PMC10359435 DOI: 10.3389/fmicb.2023.1212582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/15/2023] [Indexed: 07/25/2023] Open
Abstract
Norovirus infection is a leading cause of acute gastroenteritis worldwide and can also cause harmful chronic infections in individuals with weakened immune systems. The role of the gut microbiota in the interactions between the host and noroviruses has been extensively studied. While most past studies were conducted in vitro or focused on murine noroviruses, recent research has expanded to human noroviruses using in vivo or ex vivo human intestinal enteroids culture studies. The gut microbiota has been observed to have both promoting and inhibiting effects on human noroviruses. Understanding the interaction between noroviruses and the gut microbiota or probiotics is crucial for studying the pathogenesis of norovirus infection and its potential implications, including probiotics and vaccines for infection control. Recently, several clinical trials of probiotics and norovirus vaccines have also been published. Therefore, in this review, we discuss the current understanding and recent updates on the interactions between noroviruses and gut microbiota, including the impact of norovirus on the microbiota profile, pro-viral and antiviral effects of microbiota on norovirus infection, the use of probiotics for treating norovirus infections, and human norovirus vaccine development.
Collapse
Affiliation(s)
- Geng-Hao Bai
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Meng-Chen Tsai
- Department of General Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Sheng-Chieh Lin
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Pediatrics, Division of Allergy, Asthma and Immunology, Shuang Ho Hospital, New Taipei, Taiwan
| | - Yi-Hsiang Hsu
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Shih-Yen Chen
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Pediatrics, Division of Pediatric Gastroenterology and Hepatology, Shuang Ho Hospital, New Taipei, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
31
|
Yang L, Zhu Y, Meng J, Zhang W, Mu W. Recent progress in fucosylated derivatives of lacto- N-tetraose and lacto- N-neotetraose. Crit Rev Food Sci Nutr 2023; 64:10384-10396. [PMID: 37341681 DOI: 10.1080/10408398.2023.2224431] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Abstract
Human milk oligosaccharides (HMOs) have attracted considerable attention owing to their unique physiological functions. Two important tetrasaccharides, lacto-N-tetraose (LNT) and lacto-N-neotetraose (LNnT), are core structures of HMOs. Their safety has been evaluated and they can be added to infant formula as functional ingredients. The fucosylated derivatives of LNT and LNnT, mainly lacto-N-fucopentaose (LNFP) I, LNFP II, LNFP III, and lacto-N-difucohexaose I, exhibit prominent physiological characteristics, including modificating the intestinal microbiota, immunomodulation, anti-bacterial activities, and antiviral infection. However, they have received lesser attention than 2'-fucosyllactose. As precursors, LNT and LNnT are connected to one or two fucosyl units through α1,2/3/4 glycosidic bonds, forming a series of compounds with complex structures. These complex fucosylated oligosaccharides can be biologically synthesized using enzymatic and cell factory approaches. This review summarizes the occurrence, physiological effects, and biosynthesis of fucosylated LNT and LNnT derivatives and their future development.
Collapse
Affiliation(s)
- Longhao Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jiawei Meng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Wenli Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
32
|
Rahman T, Sarwar PF, Potter C, Comstock SS, Klepac-Ceraj V. Role of human milk oligosaccharide metabolizing bacteria in the development of atopic dermatitis/eczema. Front Pediatr 2023; 11:1090048. [PMID: 37020647 PMCID: PMC10069630 DOI: 10.3389/fped.2023.1090048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 02/23/2023] [Indexed: 04/07/2023] Open
Abstract
Despite affecting up to 20% of infants in the United States, there is no cure for atopic dermatitis (AD), also known as eczema. Atopy usually manifests during the first six months of an infant's life and is one predictor of later allergic health problems. A diet of human milk may offer protection against developing atopic dermatitis. One milk component, human milk oligosaccharides (HMOs), plays an important role as a prebiotic in establishing the infant gut microbiome and has immunomodulatory effects on the infant immune system. The purpose of this review is to summarize the available information about bacterial members of the intestinal microbiota capable of metabolizing HMOs, the bacterial genes or metabolic products present in the intestinal tract during early life, and the relationship of these genes and metabolic products to the development of AD/eczema in infants. We find that specific HMO metabolism gene sets and the metabolites produced by HMO metabolizing bacteria may enable the protective role of human milk against the development of atopy because of interactions with the immune system. We also identify areas for additional research to further elucidate the relationship between the human milk metabolizing bacteria and atopy. Detailed metagenomic studies of the infant gut microbiota and its associated metabolomes are essential for characterizing the potential impact of human milk-feeding on the development of atopic dermatitis.
Collapse
Affiliation(s)
- Trisha Rahman
- Department of Biological Sciences, Wellesley College, Wellesley, MA, United States
| | - Prioty F. Sarwar
- Department of Biological Sciences, Wellesley College, Wellesley, MA, United States
| | - Cassie Potter
- Department of Biological Sciences, Wellesley College, Wellesley, MA, United States
| | - Sarah S. Comstock
- Department of Food Science & Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Vanja Klepac-Ceraj
- Department of Biological Sciences, Wellesley College, Wellesley, MA, United States
| |
Collapse
|
33
|
Zhao M, Meng Y, Cao B, Tong J, Liu X, Yan H, Yang H, Han H, Liang X, Chen H. A bibliometric analysis of studies on gut microbiota in attention-deficit and hyperactivity disorder from 2012 to 2021. Front Microbiol 2023; 14:1055804. [PMID: 37007507 PMCID: PMC10050751 DOI: 10.3389/fmicb.2023.1055804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/27/2023] [Indexed: 03/17/2023] Open
Abstract
BackgroundAn increasing number of studies have focused on the role of gut microbiota in the treatment of ADHD, but its related molecular mechanisms are not yet clear, and there is still room for development of studies targeting this area. This study analyzes publications from 2012 to 2021 in a comprehensive and multi-faceted visualization, with the aim of grasping the existing research profile and guiding scholars to make more in-depth studies.MethodsThe 1,677 articles and 298 review articles on gut microbiota in ADHD were retrieved from the Web of Science Core Collection. CiteSpace, VOSviewer, Microsoft Excel 2019, Scimago Graphica, Bibliometrix and Pajek metrics software were used for visualization and analysis of the included literature.ResultsOn August 3, 2022, a total of 1975 English-language articles on gut microbiota in ADHD were retrieved from Web of Science Core Collection (WoSCC) from January 2012 to December 2021, with a steady upward trend in the number of articles published in this field over the decade. The top three countries in terms of the number of articles published are the United States, China, and Spain. Meanwhile, CONSEJO SUPERIOR DE INVESTIGACIONES CIENTIFICAS CSIC, UNIV OF CALIFORNIA SYSTEM, and UDICE FRENCH RESEARCH UNIV have made significant contributions in this field. In the analysis of the published journals, PLoS One was not only the first in terms of number of articles published but also the most cited. Wang J was the most prolific author and CAPORASO JG ranked first in terms of co-cited authors. In addition, “Diet rapidly and reproducibly alters the human gut microbiome,” published by David LA et al., has the highest citation frequency in this field. The most frequently occurring keyword was “gut microbiota.”ConclusionThe results of this paper clarify the current status of research on gut microbiota in ADHD. Based on the research on the mechanism of gut microbiota in other diseases, there is reason to believe that the exploration of gut microbiota in ADHD must be increasingly mature. And the study speculates that future research may focus on “nutrition supplements,” “lipid metabolism,” and “gut brain axis.” It is imperative to promote a closer international cooperation among scholars in this field.
Collapse
Affiliation(s)
- Mingyi Zhao
- Department of Pediatrics, The Third Xiangya Hopsital, Central South University, Changsha, Hunan, China
| | - Yang Meng
- Department of Pediatrics, The Third Xiangya Hopsital, Central South University, Changsha, Hunan, China
| | - Buzi Cao
- Department of Pediatrics, The Third Xiangya Hopsital, Central South University, Changsha, Hunan, China
- Medical School, Hunan Normal University, Changsha, China
| | - Jianbin Tong
- Department of Anesthesiology, The Third Xiangya Hopsital, Central South University, Changsha, Hunan, China
- Hunan Province Key Laboratory of Brain Homeostasis, The Third Xiangya Hopsital, Central South University, Changsha, Hunan, China
| | - Xiaoying Liu
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hao Yan
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hanqi Yang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Houzhi Han
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiaobing Liang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hui Chen
- Department of Clinical Laboratory, The Third Xiangya Hopsital, Central South University, Changsha, Hunan, China
- *Correspondence: Hui Chen,
| |
Collapse
|
34
|
Trompette A, Ubags ND. Skin barrier immunology from early life to adulthood. Mucosal Immunol 2023; 16:194-207. [PMID: 36868478 DOI: 10.1016/j.mucimm.2023.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/15/2023] [Accepted: 02/21/2023] [Indexed: 03/05/2023]
Abstract
Our skin has a unique barrier function, which is imperative for the body's protection against external pathogens and environmental insults. Although interacting closely and sharing many similarities with key mucosal barrier sites, such as the gut and the lung, the skin also provides protection for internal tissues and organs and has a distinct lipid and chemical composition. Skin immunity develops over time and is influenced by a multiplicity of different factors, including lifestyle, genetics, and environmental exposures. Alterations in early life skin immune and structural development may have long-term consequences for skin health. In this review, we summarize the current knowledge on cutaneous barrier and immune development from early life to adulthood, with an overview of skin physiology and immune responses. We specifically highlight the influence of the skin microenvironment and other host intrinsic, host extrinsic (e.g. skin microbiome), and environmental factors on early life cutaneous immunity.
Collapse
Affiliation(s)
- Aurélien Trompette
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Niki D Ubags
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland.
| |
Collapse
|
35
|
Xie L, Alam MJ, Marques FZ, Mackay CR. A major mechanism for immunomodulation: Dietary fibres and acid metabolites. Semin Immunol 2023; 66:101737. [PMID: 36857894 DOI: 10.1016/j.smim.2023.101737] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 01/17/2023] [Accepted: 02/09/2023] [Indexed: 03/01/2023]
Abstract
Diet and the gut microbiota have a profound influence on physiology and health, however, mechanisms are still emerging. Here we outline several pathways that gut microbiota products, particularly short-chain fatty acids (SCFAs), use to maintain gut and immune homeostasis. Dietary fibre is fermented by the gut microbiota in the colon, and large quantities of SCFAs such as acetate, propionate, and butyrate are produced. Dietary fibre and SCFAs enhance epithelial integrity and thereby limit systemic endotoxemia. Moreover, SCFAs inhibit histone deacetylases (HDAC), and thereby affect gene transcription. SCFAs also bind to 'metabolite-sensing' G-protein coupled receptors (GPCRs) such as GPR43, which promotes immune homeostasis. The enormous amounts of SCFAs produced in the colon are sufficient to lower pH, which affects the function of proton sensors such as GPR65 expressed on the gut epithelium and immune cells. GPR65 is an anti-inflammatory Gαs-coupled receptor, which leads to the inhibition of inflammatory cytokines. The importance of GPR65 in inflammatory diseases is underscored by genetics associated with the missense variant I231L (rs3742704), which is associated with human inflammatory bowel disease, atopic dermatitis, and asthma. There is enormous scope to manipulate these pathways using specialized diets that release very high amounts of specific SCFAs in the gut, and we believe that therapies that rely on chemically modified foods is a promising approach. Such an approach includes high SCFA-producing diets, which we have shown to decrease numerous inflammatory western diseases in mouse models. These diets operate at many levels - increased gut integrity, changes to the gut microbiome, and promotion of immune homeostasis, which represents a new and highly promising way to prevent or treat human disease.
Collapse
Affiliation(s)
- Liang Xie
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Hypertension Research Laboratory, School of Biological Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Md Jahangir Alam
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Clayton, VIC 3800, Australia; Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne,VIC 3004, Australia
| | - Charles R Mackay
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China.
| |
Collapse
|
36
|
Hu M, Li M, Li C, Miao M, Zhang T. Effects of Human Milk Oligosaccharides in Infant Health Based on Gut Microbiota Alteration. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:994-1001. [PMID: 36602115 DOI: 10.1021/acs.jafc.2c05734] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The primary active components of breast milk are human milk oligosaccharides (HMOs). HMOs provide many benefits to infants, including regulating their metabolism, immune system, and brain development. Recent studies have emphasized that HMOs act as prebiotics by the metabolism of intestinal microorganisms to produce short-chain fatty acids, which are crucial for infant development. In addition, HMOs with different structural characteristics can form different microbial compositions. HMOs-induced predominant microbes, including Bifidobacterium infantis, B. bifidum, B. breve, and B. longum, and their metabolites demonstrated pertinent health-promoting properties. Meanwhile, HMOs could also directly reduce the occurrence of diseases through the effects of preventing pathogen infection. In this review, we address the probable function of HMOs inside the HMOs-gut microbiota-infant network, by describing the physiological functions of HMOs and the implications of diet on the HMOs-gut microbiota-infant network.
Collapse
Affiliation(s)
- Miaomiao Hu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Mengli Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Chenchen Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ming Miao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Tao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
37
|
Abstract
Human milk oligosaccharides (HMOs) are the third most important solid component in human milk and act in tandem with other bioactive components. Individual HMO levels and distribution vary greatly between mothers by multiple variables, such as secretor status, race, geographic region, environmental conditions, season, maternal diet, and weight, gestational age and mode of delivery. HMOs improve the gastrointestinal barrier and also promote a bifidobacterium-rich gut microbiome, which protects against infection, strengthens the epithelial barrier, and creates immunomodulatory metabolites. HMOs fulfil a variety of physiologic functions including potential support to the immune system, brain development, and cognitive function. Supplementing infant formula with HMOs is safe and promotes a healthy development of the infant revealing benefits for microbiota composition and infection prevention. Because of limited data comparing the effect of non-human oligosaccharides to HMOs, it is not known if HMOs offer an additional clinical benefit over non-human oligosaccharides. Better knowledge of the factors influencing HMO composition and their functions will help to understand their short- and long-term benefits.
Collapse
Affiliation(s)
- Meltem Dinleyici
- Department of Social Pediatrics, Eskisehir Osmangazi University Faculty of Medicine, Eskisehir, Turkey
| | - Jana Barbieur
- UZ Brussel, KidZ Health Castle, Vrije Unversiteit Brussel, Brussels, Belgium
| | - Ener Cagri Dinleyici
- Department of Pediatrics, Eskisehir Osmangazi University Faculty of Medicine, Eskisehir, Turkey
| | - Yvan Vandenplas
- UZ Brussel, KidZ Health Castle, Vrije Unversiteit Brussel, Brussels, Belgium
| |
Collapse
|
38
|
Fibbiani M, Ghelli Luserna DI Rorà L, Novelli T, Peroni DG. The impact of human milk oligosaccharides on health from infancy to childhood. Minerva Pediatr (Torino) 2022; 74:724-732. [PMID: 36178339 DOI: 10.23736/s2724-5276.22.07037-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Human milk oligosaccharides (HMOs) act as prebiotics in the infant's gut and contribute to the relationship among the host and the gut microbiota. HMO are greatly present in the human milk and their benefit may include: reinforcement of the immune system with a better immune response to infective agents, improved resistance to infections of the gut, immunomodulation against food allergies, asthma, and atopic dermatitis and finally decreased the risk of chronic diseases. In this narrative review will discuss evidence present in literature regarding HMOs in human milk and their supplementation in infant formula.
Collapse
Affiliation(s)
- Martina Fibbiani
- Section of Pediatrics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Tommaso Novelli
- Section of Pediatrics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Diego G Peroni
- Section of Pediatrics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy -
| |
Collapse
|
39
|
Trbojević-Akmačić I, Lageveen-Kammeijer GSM, Heijs B, Petrović T, Deriš H, Wuhrer M, Lauc G. High-Throughput Glycomic Methods. Chem Rev 2022; 122:15865-15913. [PMID: 35797639 PMCID: PMC9614987 DOI: 10.1021/acs.chemrev.1c01031] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Glycomics aims to identify the structure and function of the glycome, the complete set of oligosaccharides (glycans), produced in a given cell or organism, as well as to identify genes and other factors that govern glycosylation. This challenging endeavor requires highly robust, sensitive, and potentially automatable analytical technologies for the analysis of hundreds or thousands of glycomes in a timely manner (termed high-throughput glycomics). This review provides a historic overview as well as highlights recent developments and challenges of glycomic profiling by the most prominent high-throughput glycomic approaches, with N-glycosylation analysis as the focal point. It describes the current state-of-the-art regarding levels of characterization and most widely used technologies, selected applications of high-throughput glycomics in deciphering glycosylation process in healthy and disease states, as well as future perspectives.
Collapse
Affiliation(s)
| | | | - Bram Heijs
- Center
for Proteomics and Metabolomics, Leiden
University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Tea Petrović
- Genos,
Glycoscience Research Laboratory, Borongajska cesta 83H, 10 000 Zagreb, Croatia
| | - Helena Deriš
- Genos,
Glycoscience Research Laboratory, Borongajska cesta 83H, 10 000 Zagreb, Croatia
| | - Manfred Wuhrer
- Center
for Proteomics and Metabolomics, Leiden
University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Gordan Lauc
- Genos,
Glycoscience Research Laboratory, Borongajska cesta 83H, 10 000 Zagreb, Croatia
- Faculty
of Pharmacy and Biochemistry, University
of Zagreb, A. Kovačića 1, 10 000 Zagreb, Croatia
| |
Collapse
|
40
|
Dynamics of human milk oligosaccharides in early lactation and relation with growth and appetitive traits of Filipino breastfed infants. Sci Rep 2022; 12:17304. [PMID: 36243744 PMCID: PMC9569346 DOI: 10.1038/s41598-022-22244-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/12/2022] [Indexed: 01/10/2023] Open
Abstract
Human milk oligosaccharides play a key role in the maturation of the infant gut microbiome and immune system and are hypothesized to affect growth. This study examined the temporal changes of 24 HMOs and their associations to infant growth and appetitive traits in an exploratory, prospective, observational, study of 41 Filipino mother-infant dyads. Exclusively breastfed, healthy, term infants were enrolled at 21-26 days of age (≈ 0.75 mo) and followed for 6 months. Infant growth measures and appetitive traits were collected at visit 1 (V1) (≈ 0.75 mo), V2 (≈ 1.5 mo), V3 (2.5 mo), V4 (2.75 mo), V5 (4 mo), and V6 (6 mo), while HMOs were measured at V1, V2, V3 and V5. Overall exposure to each HMO was summarized as area under the curve from baseline to 4 months of age and examined in association with each measure of growth at 6 months using linear regression adjusted for maternal age at birth, infant sex, birth weight, and mode of delivery. We saw modest associations between several HMOs and infant growth parameters. Our results suggest that specific HMOs, partly as proxy for milk groups (defined by Secretor and Lewis status), may be associated with head circumference and length, increasing their relevance especially in populations at the lower end of the WHO growth curve. We did not identify the same HMOs associated with infant appetitive traits, indicating that at least in our cohort, changes in appetite were not driving the observed associations between HMOs and growth.Clinical trial registration: NCT03387124.
Collapse
|
41
|
Pivrncova E, Kotaskova I, Thon V. Neonatal Diet and Gut Microbiome Development After C-Section During the First Three Months After Birth: A Systematic Review. Front Nutr 2022; 9:941549. [PMID: 35967823 PMCID: PMC9364824 DOI: 10.3389/fnut.2022.941549] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/15/2022] [Indexed: 11/28/2022] Open
Abstract
Background Cesarean section (C-section) delivery imprints fundamentally on the gut microbiota composition with potential health consequences. With the increasing incidence of C-sections worldwide, there is a need for precise characterization of neonatal gut microbiota to understand how to restore microbial imbalance after C-section. After birth, gut microbiota development is shaped by various factors, especially the infant’s diet and antibiotic exposure. Concerning diet, current research has proposed that breastfeeding can restore the characteristic gut microbiome after C-section. Objectives In this systematic review, we provide a comprehensive summary of the current literature on the effect of breastfeeding on gut microbiota development after C-section delivery in the first 3 months of life. Methods The retrieved data from PubMed, Scopus, and Web of Science were evaluated according to the PICO/PECO strategy. Quality assessment was conducted by the Newcastle–Ottawa Scale. Results After critical selection, we identified 14 out of 4,628 studies for the evaluation of the impact of the diet after C-section delivery. The results demonstrate consistent evidence that C-section and affiliated intrapartum antibiotic exposure affect Bacteroidetes abundance and the incapacity of breastfeeding to reverse their reduction. Furthermore, exclusive breastfeeding shows a positive effect on Actinobacteria and Bifidobacteria restoration over the 3 months after birth. None of the included studies detected any significant changes in Lactobacillus abundance in breastfed infants after C-section. Conclusion C-section and intrapartum antibiotic exposure influence an infant’s gut microbiota by depletion of Bacteroides, regardless of the infant’s diet in the first 3 months of life. Even though breastfeeding increases the presence of Bifidobacteria, further research with proper feeding classification is needed to prove the restoration effect on some taxa in infants after C-section. Systematic Review Registration: [www.crd.york.ac.uk/prospero/], identifier [CRD42021287672].
Collapse
Affiliation(s)
- Eliska Pivrncova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Iva Kotaskova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Vojtech Thon
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| |
Collapse
|
42
|
Bosheva M, Tokodi I, Krasnow A, Pedersen HK, Lukjancenko O, Eklund AC, Grathwohl D, Sprenger N, Berger B, Cercamondi CI. Infant Formula With a Specific Blend of Five Human Milk Oligosaccharides Drives the Gut Microbiota Development and Improves Gut Maturation Markers: A Randomized Controlled Trial. Front Nutr 2022; 9:920362. [PMID: 35873420 PMCID: PMC9298649 DOI: 10.3389/fnut.2022.920362] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/27/2022] [Indexed: 11/17/2022] Open
Abstract
Background Human milk oligosaccharides (HMOs) have important biological functions for a healthy development in early life. Objective This study aimed to investigate gut maturation effects of an infant formula containing five HMOs (2′-fucosyllactose, 2′,3-di-fucosyllactose, lacto-N-tetraose, 3′-sialyllactose, and 6′-sialyllactose). Methods In a multicenter study, healthy infants (7–21 days old) were randomly assigned to a standard cow’s milk-based infant formula (control group, CG); the same formula with 1.5 g/L HMOs (test group 1, TG1); or with 2.5 g/L HMOs (test group 2, TG2). A human milk-fed group (HMG) was enrolled as a reference. Fecal samples collected at baseline (n∼150/formula group; HMG n = 60), age 3 (n∼140/formula group; HMG n = 65) and 6 (n∼115/formula group; HMG n = 60) months were analyzed for microbiome (shotgun metagenomics), metabolism, and biomarkers. Results At both post-baseline visits, weighted UniFrac analysis indicated different microbiota compositions in the two test groups (TGs) compared to CG (P < 0.01) with coordinates closer to that of HMG. The relative abundance of Bifidobacterium longum subsp. infantis (B. infantis) was higher in TGs vs. CG (P < 0.05; except at 6 months: TG2 vs. CG P = 0.083). Bifidobacterium abundance was higher by ∼45% in TGs vs. CG at 6-month approaching HMG. At both post-baseline visits, toxigenic Clostridioides difficile abundance was 75–85% lower in TGs vs. CG (P < 0.05) and comparable with HMG. Fecal pH was significantly lower in TGs vs. CG, and the overall organic acid profile was different in TGs vs. CG, approaching HMG. At 3 months, TGs (vs. CG) had higher secretory immunoglobulin A (sIgA) and lower alpha-1-antitrypsin (P < 0.05). At 6 months, sIgA in TG2 vs. CG remained higher (P < 0.05), and calprotectin was lower in TG1 (P < 0.05) vs. CG. Conclusion Infant formula with a specific blend of five HMOs supports the development of the intestinal immune system and gut barrier function and shifts the gut microbiome closer to that of breastfed infants with higher bifidobacteria, particularly B. infantis, and lower toxigenic Clostridioides difficile. Clinical Trial Registration [https://clinicaltrials.gov/ct2/show/], identifier [NCT03722550].
Collapse
Affiliation(s)
- Miroslava Bosheva
- University Multiprofile Hospital for Active Treatment, St. George Medical University, Plovdiv, Bulgaria
| | - Istvan Tokodi
- Infant and Children’s Department, St. George’s Hospital, Székesfehérvár, Hungary
| | | | | | | | | | | | - Norbert Sprenger
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Bernard Berger
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
- *Correspondence: Bernard Berger,
| | - Colin I. Cercamondi
- Nestlé Product Technology Center – Nutrition, Société des Produits Nestlé S.A., Vevey, Switzerland
| | - 5 HMO Study Investigator ConsortiumBauerViktorArciszewskaMalgorzataTarnevaMariaPopovaIrinaDosevSvilenDimitrovaSirmaNikolovaOlgaNowakMarzenaSzuflinska-SidorowiczMagdalenaKorczowskiBartoszKarcheva-BeloevaRositsaBanovStefanCimoszkoBoguslawaOlechowskiWieslawSimkoRobertTengelyiZsuzsannaKorbalPiotrZolnowskaMartaBilevAntonVasilopoulosGeorgiosKorzynskaSylwiaLakiIstvánKoleva-SyarovaMargaritaGrigorovToniKraevaSteliyanaKovácsÉvaMarkovaRadaJasieniak-PinisGrazynaFisterKatalinStoevaTatyanaDr. Kenessey Albert Hospital and Clinic, Balassagyarmat, Hungary; Polyclinic of Gynecology and Obstetrics Arciszewscy, Bialystok, Poland; University Multiprofile Hospital for Active Treatment Deva Mariya—Neonatology, Burgas, Bulgaria; Medical Center Prolet—Pediatrics department, Ruse, Bulgaria; Medical Center Excelsior, Sofia, Bulgaria; Multiprofile Hospital for Active Treatment Sveti Ivan Rilski, Kozloduy, Bulgaria; Medical Center PROMED, Krakow, Poland; Medical Center Pratia Warszawa, Warszawa, Poland; College of Medical Sciences, University of Rzeszów, Rzeszów, Poland; Medical Center-1, Sevlievo, Bulgaria; Individual Practice for Specialized Medical Assistance, Stara Zagora, Bulgaria; Primary Health Care Clinic Clinical Vitae, Gdansk, Poland; ALERGO-MED Specialist Medical Clinic, Tarnow, Poland; Futurenest Clinical Research, Miskolc, Hungary; Medical Center Clinexpert, Budapest, Hungary; Dr. Jan Biziel’s University Hospital No. 2, Bydgoszcz, Poland; Plejady Medical Center, Krakow, Poland; Medical Center Sveti Ivan Rilski Chudotvorets, Blagoevgrad, Bulgaria; Center of Innovative Therapies, Piaseczno, Poland; Medical Center Pratia Ostroleka, Ostroleka, Poland; Kanizsai Dorottya Hospital, Nagykanizsa, Hungary; Diagnostic Consultative Center Ritam, Stara Zagora, Bulgaria; Multiprofile Hospital for Active Treatment Sveti Georgi, Montana, Bulgaria; Alitera Medical Centre, Sofia, Bulgaria; Family Pediatric Surgery/Babadoki Ltd., Szeged, Hungary; Policlinic Bulgaria—Department of pediatrics; Sofia, Bulgaria; Non-public Health Care Institution Specialist Clinics ATOPIA, Krakow, Poland; Bugát Pál Hospital—Department of Pediatrics, Gyöngyös, Hungary; Medical Center—Izgrev Ltd., Sofia, Bulgaria.
| |
Collapse
|
43
|
Park JS. Clinical importance of immunonutrition in infants: a review of the recent literature. Clin Exp Pediatr 2022; 65:337-343. [PMID: 35176834 PMCID: PMC9263427 DOI: 10.3345/cep.2021.00570] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 12/15/2021] [Indexed: 12/04/2022] Open
Abstract
During the last decades, the role of nutrition has been well elucidated in medicine, especially among critically ill infants and children. Many nutrients have the potential to modulate the immune system. A healthy immune system is essential for the prevention and recovery of many pediatric illnesses. Intervention using specific nutrients for the immune system is called immunonutrition. Immunonutrient supplementation has been attempted to modulate inflammatory or immune responses, leading to an improved clinical course of critically ill patients with prolonged nutritional supplementation parenterally or enterally. This review discusses immunomodulatory nutrients for infants based on the recent literature.
Collapse
Affiliation(s)
- Ji Sook Park
- Department of Pediatrics, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, Korea.,Institute of Health Sciences, Gyeongsang National University, Jinju, Korea
| |
Collapse
|
44
|
Białek-Dratwa A, Soczewka M, Grajek M, Szczepańska E, Kowalski O. Use of the Baby-Led Weaning (BLW) Method in Complementary Feeding of the Infant-A Cross-Sectional Study of Mothers Using and Not Using the BLW Method. Nutrients 2022; 14:nu14122372. [PMID: 35745102 PMCID: PMC9227137 DOI: 10.3390/nu14122372] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 02/01/2023] Open
Abstract
Baby-led weaning (BLW) is an increasingly popular way of expanding a baby’s diet. It is based on the baby becoming physically ready to feed himself, effectively supplementing his diet, which until now has been based on breast milk or modified milk. The aim of the study was to assess mothers’ knowledge about the use of the BLW method to expand the diet of a young child. The essence of the study assumed the analysis of the advantages and disadvantages of using this method indicated by mothers. Materials and Methods: A total of 320 mothers participated in the study. Data for the study were collected anonymously using the CAWI method. The research tool was the original questionnaire relating to the knowledge about the BLW method and the application of the BLW method in practice. Results: The BLW method was used by 240 (75%) women. The reasons for not using the BLW method were: the child did not cooperate n = 30 (37.5%) and was not ready to use the BLW method n = 20 (25%). In total, 182 (75.8%) mothers using BLW and 63 (78.8%) mothers not using BLW started extending the diet before the child was 6 months old. According to 270 (84.4%) mothers, including 205 (85.4%) using BLW, stable sitting in a highchair/on the lap is a decisive factor for starting the dietary expansion with the BLW method. Conclusions: Mothers’ knowledge of the BLW method as a way of expanding a young child’s diet was insufficient. It seems important to implement appropriate educational activities on the methods of expanding children’s diets to broaden parents’ knowledge of the influence of nutrition on infant development.
Collapse
Affiliation(s)
- Agnieszka Białek-Dratwa
- Department of Human Nutrition, Department of Dietetics, Faculty of Health Sciences in Bytom, Medical University of Silesia in Katowice, ul. Jordana 19, 41-808 Zabrze, Poland; (E.S.); (O.K.)
- Correspondence: ; Tel.: +48-(0-32)-275-51-95
| | - Monika Soczewka
- Department of Pediatric Diabetes, Auxology and Obesity, Poznan University of Medical Sciences, 60-512 Poznan, Poland;
- Doctoral School, Poznan University of Medical Sciences, Fredry St. 10, 61-701 Poznan, Poland
| | - Mateusz Grajek
- Department of Public Health, Department of Public Health Policy, Faculty of Health Sciences in Bytom, Medical University of Silesia in Katowice, ul. Piekarska 18, 41-902 Bytom, Poland;
| | - Elżbieta Szczepańska
- Department of Human Nutrition, Department of Dietetics, Faculty of Health Sciences in Bytom, Medical University of Silesia in Katowice, ul. Jordana 19, 41-808 Zabrze, Poland; (E.S.); (O.K.)
| | - Oskar Kowalski
- Department of Human Nutrition, Department of Dietetics, Faculty of Health Sciences in Bytom, Medical University of Silesia in Katowice, ul. Jordana 19, 41-808 Zabrze, Poland; (E.S.); (O.K.)
| |
Collapse
|
45
|
Manipulating Microbiota to Treat Atopic Dermatitis: Functions and Therapies. Pathogens 2022; 11:pathogens11060642. [PMID: 35745496 PMCID: PMC9228373 DOI: 10.3390/pathogens11060642] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/27/2022] [Accepted: 05/29/2022] [Indexed: 12/13/2022] Open
Abstract
Atopic dermatitis (AD) is a globally prevalent skin inflammation with a particular impact on children. Current therapies for AD are challenged by the limited armamentarium and the high heterogeneity of the disease. A novel promising therapeutic target for AD is the microbiota. Numerous studies have highlighted the involvement of the skin and gut microbiota in the pathogenesis of AD. The resident microbiota at these two epithelial tissues can modulate skin barrier functions and host immune responses, thus regulating AD progression. For example, the pathogenic roles of Staphylococcus aureus in the skin are well-established, making this bacterium an attractive target for AD treatment. Targeting the gut microbiota is another therapeutic strategy for AD. Multiple oral supplements with prebiotics, probiotics, postbiotics, and synbiotics have demonstrated promising efficacy in both AD prevention and treatment. In this review, we summarize the association of microbiota dysbiosis in both the skin and gut with AD, and the current knowledge of the functions of commensal microbiota in AD pathogenesis. Furthermore, we discuss the existing therapies in manipulating both the skin and gut commensal microbiota to prevent or treat AD. We also propose potential novel therapies based on the cutting-edge progress in this area.
Collapse
|
46
|
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: 4.7] [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
|
47
|
Bai Y, Yang X, Yu H, Chen X. Substrate and Process Engineering for Biocatalytic Synthesis and Facile Purification of Human Milk Oligosaccharides. CHEMSUSCHEM 2022; 15:e202102539. [PMID: 35100486 PMCID: PMC9272545 DOI: 10.1002/cssc.202102539] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/30/2022] [Indexed: 05/08/2023]
Abstract
Innovation in process development is essential for applying biocatalysis in industrial and laboratory production of organic compounds, including beneficial carbohydrates such as human milk oligosaccharides (HMOs). HMOs have attracted increasing attention for their potential application as key ingredients in products that can improve human health. To efficiently access HMOs through biocatalysis, a combined substrate and process engineering strategy is developed, namely multistep one-pot multienzyme (MSOPME) design. The strategy allows access to a pure tagged HMO in a single reactor with a single C18-cartridge purification process, despite the length of the target. Its efficiency is demonstrated in the high-yielding (71-91 %) one-pot synthesis of twenty tagged HMOs (83-155 mg), including long-chain oligosaccharides with or without fucosylation or sialylation up to nonaoses from a lactoside without the isolation of the intermediate oligosaccharides. Gram-scale synthesis of an important HMO derivative - tagged lacto-N-fucopentaose-I (LNFP-I) - proceeds in 84 % yield. Tag removal is carried out in high efficiency (94-97 %) without the need for column purification to produce the desired natural HMOs with a free reducing end. The method can be readily adapted for large-scale synthesis and automation to allow quick access to HMOs, other glycans, and glycoconjugates.
Collapse
Affiliation(s)
- Yuanyuan Bai
- Department of Chemistry, University of California, Davis, One Shields Avenue, 95616, Davis, California, USA
| | - Xiaohong Yang
- Department of Chemistry, University of California, Davis, One Shields Avenue, 95616, Davis, California, USA
| | - Hai Yu
- Department of Chemistry, University of California, Davis, One Shields Avenue, 95616, Davis, California, USA
| | - Xi Chen
- Department of Chemistry, University of California, Davis, One Shields Avenue, 95616, Davis, California, USA
| |
Collapse
|
48
|
Phipps KR, Lozon D, Stannard DR, Gilby B, Baldwin N, Mikš MH, Lau A, Röhrig CH. Neonatal subchronic toxicity and in vitro genotoxicity studies of the human-identical milk oligosaccharide 3-fucosyllactose. J Appl Toxicol 2022; 42:1671-1687. [PMID: 35510931 DOI: 10.1002/jat.4335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/01/2022] [Indexed: 11/10/2022]
Abstract
Human milk oligosaccharides, such as 3-fucosyllactose (3-FL), are bioactive components of breast milk associated with benefits for infant growth and development. Structurally identical compounds (human-identical milk oligosaccharides - HiMOs) can be produced using microbial fermentation, allowing their use in infant formula to increase its similarity with human milk. Toxicological studies are required to demonstrate safety of HiMOs and that of any impurities potentially carried over from the manufacturing process. Biotechnologically produced 3-FL was tested for potential genotoxicity (bacterial reverse mutation test and in vitro mammalian micronucleus test) and subchronic toxicity (90-day study with neonatal rats). In the 90-day study, 3-FL was administered by gavage to rats once daily from Day 7 of age, at doses up to 4000 mg/kg body weight (bw)/day (the maximum feasible dose), followed by a 4-week recovery period. Reference controls received 4000 mg/kg bw/day of oligofructose, an ingredient permitted for use in infant formula. Results for the genotoxicity studies were negative. In the 90-day study, there were no adverse effects of 3-FL on any of the parameters measured; thus, the no-observed-adverse-effect level was 4000 mg/kg bw/day (the highest dose tested). These results support the safety of biotechnologically produced 3-FL for use in infant formula and other foods.
Collapse
Affiliation(s)
| | - Dayna Lozon
- Intertek Health Sciences Inc., Mississauga, Ontario, Canada
| | - Diane R Stannard
- Labcorp Early Development Laboratories Limited (formerly Covance Laboratories Limited), Eye, Suffolk, UK
| | - Ben Gilby
- Labcorp Early Development Laboratories Limited (formerly Covance Laboratories Limited), Woolley Road, Alconbury, Huntingdon, Cambridgeshire, UK
| | | | - Marta Hanna Mikš
- Glycom A/S, Hørsholm, Denmark.,University of Warmia and Mazury in Olsztyn, Faculty of Food Science, Olsztyn, Poland
| | | | | |
Collapse
|
49
|
Pandey A, Gupta VK. Special Issue 'Microbial glycobiotechnology'. Microb Cell Fact 2022; 21:54. [PMID: 35392921 PMCID: PMC8991797 DOI: 10.1186/s12934-022-01784-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Ashok Pandey
- Centre for Innovation and Translational Research, CSIR-Indian Institute for Toxicology Research (CSIR-IITR), Lucknow, 226001, India
- Centre for Energy and Environmental Sustainability, Lucknow, 226029, Uttar Pradesh, India
- Sustainability Cluster, School of Engineering, University of Petroleum and Energy Studies, Dehradun, 248007, Uttarkhand, India
| | - Vijai Kumar Gupta
- Biorefining and Advanced Materials Research Center, SRUC, Kings Buildings, West Mains Road, Edinburgh, EH9 3JG, UK.
- Center for Safe and Improved Food, SRUC, Kings Buildings, West Mains Road, Edinburgh, EH9 3JG, UK.
| |
Collapse
|
50
|
Lundstrøm J, Bojar D. Structural insights into host-microbe glycointeractions. Curr Opin Struct Biol 2022; 73:102337. [PMID: 35182928 DOI: 10.1016/j.sbi.2022.102337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/30/2021] [Accepted: 01/14/2022] [Indexed: 11/03/2022]
Abstract
Despite their ubiquitous presence in biological systems, glycans have historically received less attention than they deserved. Investigations in recent years have featured important findings about the role of glycans in regulating the human gut microbiota. Here, we present a brief overview of current trends that shape future directions of computational and experimental research approaches and add to our understanding of host-microbe glycointeractions.
Collapse
Affiliation(s)
- Jon Lundstrøm
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden. https://twitter.com/jonlundstrm
| | - Daniel Bojar
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|