1
|
Natal ACDC, de Paula Menezes R, de Brito Röder DVD. Role of maternal milk in providing a healthy intestinal microbiome for the preterm neonate. Pediatr Res 2024:10.1038/s41390-024-03751-x. [PMID: 39663425 DOI: 10.1038/s41390-024-03751-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 10/28/2024] [Accepted: 11/10/2024] [Indexed: 12/13/2024]
Abstract
The immature gastrointestinal tract of preterm neonates leads to a delayed and distinctive establishment of the gut microbiome, making them susceptible to potentially pathogenic bacteria and increasing the risk of infections. Maternal milk, recognized as the optimal source of nutrition, plays a multifaceted role in modulating the gut microbiome of premature newborns. Human milk oligosaccharides, acting as prebiotics, provide essential nourishment for key bacteria such as Bifidobacterium, contributing to the proliferation of beneficial bacterial populations. Additionally, maternal milk is rich in Immunoglobulins that stimulate immune cell responses, providing protective effects on the infant's gut mucosa. Moreover, bioactive proteins such as secretory immunoglobulin A (SIgA), lactoferrin, lysozyme, and mucins play a crucial role in defending against pathogens and regulating the immune system at the cellular level. These proteins contribute not only to infection prevention but also emphasize the impact of breast milk in fortifying the body's innate defenses. This multifaceted role of maternal milk, including essential nutrients, beneficial bacteria, and bioactive proteins, highlights the importance of promoting the mother's own milk feeding in the Neonatal Intensive Care Unit (NICU). It not only optimizes the long-term outcomes and well-being of preterm infants but also provides a holistic approach to their health and development. IMPACT: This article contributes to the current understanding of the relationship between breastfeeding and the intestinal microbiota. Fill gaps in existing literature about the subject. Provides new insights for future research.
Collapse
Affiliation(s)
- Ana Catarina de Castro Natal
- Undergraduate Nursing, Faculty of Medicine (FAMED), Federal University of Uberlandia UFU, Uberlandia, MG, Brazil.
| | | | | |
Collapse
|
2
|
Froń A, Orczyk-Pawiłowicz M. Breastfeeding Beyond Six Months: Evidence of Child Health Benefits. Nutrients 2024; 16:3891. [PMID: 39599677 PMCID: PMC11597163 DOI: 10.3390/nu16223891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Breastfeeding is globally recognized as the optimal method of infant nutrition, offering health benefits for both the child and the mother, making it a public health priority. However, the potential advantages of breastfeeding extend well beyond initial months. Breast milk adapts to the evolving needs of the growing infant, and its immunological, microbiological, and biochemical properties have been associated with enhanced protection against infections and chronic diseases, improved growth and development, and lower rates of hospitalization and mortality. This review explores the evidence supporting the continuation of breastfeeding beyond six months. More meticulous studies employing consistent methodologies and addressing confounders are essential. This will enable a more accurate determination of the extent and mechanisms of the positive impact of prolonged breastfeeding and allow for the implementation of effective public health strategies.
Collapse
Affiliation(s)
- Anita Froń
- Division of Chemistry and Immunochemistry, Department of Biochemistry and Immunochemistry, Wroclaw Medical University, M. Skłodowskiej-Curie 48/50, 50-369 Wroclaw, Poland;
| | | |
Collapse
|
3
|
Thacker N, Duncanson K, Eslick GD, Dutt S, O'Loughlin EV, Hoedt EC, Collins CE. Antibiotics, passive smoking, high socioeconomic status and sweetened foods contribute to the risk of paediatric inflammatory bowel disease: A systematic review with meta-analysis. J Pediatr Gastroenterol Nutr 2024; 79:610-621. [PMID: 39020449 DOI: 10.1002/jpn3.12303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 05/27/2024] [Accepted: 06/05/2024] [Indexed: 07/19/2024]
Abstract
OBJECTIVE Genetic and environmental factors influence pathogenesis and rising incidence of paediatric inflammatory bowel disease (PIBD). The aim was to meta-analyse evidence of diet and environmental factors in PIBD. METHODS A systematic search was conducted to identify diet and environmental factors with comparable risk outcome measures and had been reported in two or more PIBD studies for inclusion in meta-analyses. Those with ≥2 PIBD risk estimates were combined to provide pooled risk estimates. RESULTS Of 4763 studies identified, 36 studies were included. PIBD was associated with higher risk with exposure to ≥/=4 antibiotic courses (includes prescriptions/purchases/courses), passive smoking, not being breastfed, sugary drink intake, being a non-Caucasian child living in a high-income country and infection history (odds ratio [OR] range: 2-3.8). Paediatric Crohn's disease (CD) was associated with higher risk with exposure to antibiotics during early childhood, ≥/=4 antibiotic courses, high socioeconomic status (SES), maternal smoking, history of atopic conditions and infection history (OR range: 1.6-4.4). A history of infection was also associated with higher risk of paediatric ulcerative colitis (UC) (OR: 3.73). Having a higher number of siblings (≥2) was associated with lower risk of paediatric CD (OR: 0.6) and paediatric UC (OR: 0.7). Pet exposure was associated with lower risk of paediatric UC (OR: 0.5). CONCLUSION Several factors associated with PIBD risk were identified that could potentially be used to develop a disease screening tool. Future research is needed to address risk reduction in PIBD.
Collapse
Affiliation(s)
- Nisha Thacker
- School of Health Sciences, College of Health Medicine and Wellbeing, The University of Newcastle, Sydney, New South Wales, Australia
- Food and Nutrition Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Kerith Duncanson
- Food and Nutrition Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- School of Medicine and Public Health, College of Health Medicine and Wellbeing, The University of Newcastle, Sydney, New South Wales, Australia
- NHMRC Centre of Research Excellence in Digestive Health, The University of Newcastle, Sydney, New South Wales, Australia
| | - Guy D Eslick
- NHMRC Centre of Research Excellence in Digestive Health, The University of Newcastle, Sydney, New South Wales, Australia
| | - Shoma Dutt
- Department of Gastroenterology, The Children's Hospital at Westmead, Sydney Children's Hospital Network, Westmead, New South Wales, Australia
- Children's Hospital at Westmead Clinical School, Sydney Medical Program, University of Sydney, Sydney, New South Wales, Australia
| | - Edward V O'Loughlin
- Department of Gastroenterology, The Children's Hospital at Westmead, Sydney Children's Hospital Network, Westmead, New South Wales, Australia
| | - Emily C Hoedt
- NHMRC Centre of Research Excellence in Digestive Health, The University of Newcastle, Sydney, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, College of Health Medicine and Wellbeing, The University of Newcastle, Sydney, New South Wales, Australia
| | - Clare E Collins
- School of Health Sciences, College of Health Medicine and Wellbeing, The University of Newcastle, Sydney, New South Wales, Australia
- Food and Nutrition Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
4
|
Fagan BT, Constable GWA, Law R. Maternal transmission as a microbial symbiont sieve, and the absence of lactation in male mammals. Nat Commun 2024; 15:5341. [PMID: 38937464 PMCID: PMC11211401 DOI: 10.1038/s41467-024-49559-5] [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: 12/21/2022] [Accepted: 06/11/2024] [Indexed: 06/29/2024] Open
Abstract
Gut microbiomes of mammals carry a complex symbiotic assemblage of microorganisms. Feeding newborn infants milk from the mammary gland allows vertical transmission of the parental milk microbiome to the offspring's gut microbiome. This has benefits, but also has hazards for the host population. Using mathematical models, we demonstrate that biparental vertical transmission enables deleterious microbial elements to invade host populations. In contrast, uniparental vertical transmission acts as a sieve, preventing these invasions. Moreover, we show that deleterious symbionts generate selection on host modifier genes that keep uniparental transmission in place. Since microbial transmission occurs during birth in placental mammals, subsequent transmission of the milk microbiome needs to be maternal to avoid the spread of deleterious elements. This paper therefore argues that viviparity and the hazards from biparental transmission of the milk microbiome, together generate selection against male lactation in placental mammals.
Collapse
Affiliation(s)
- Brennen T Fagan
- Leverhulme Centre for Anthropocene Biodiversity, University of York, York, UK.
- Department of Mathematics, University of York, York, UK.
| | | | - Richard Law
- Department of Mathematics, University of York, York, UK
| |
Collapse
|
5
|
Pinckard KM, Félix-Soriano E, Hamilton S, Terentyeva R, Baer LA, Wright KR, Nassal D, Esteves JV, Abay E, Shettigar VK, Ziolo MT, Hund TJ, Wold LE, Terentyev D, Stanford KI. Maternal exercise preserves offspring cardiovascular health via oxidative regulation of the ryanodine receptor. Mol Metab 2024; 82:101914. [PMID: 38479548 PMCID: PMC10965826 DOI: 10.1016/j.molmet.2024.101914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 03/19/2024] Open
Abstract
OBJECTIVE The intrauterine environment during pregnancy is a critical factor in the development of obesity, diabetes, and cardiovascular disease in offspring. Maternal exercise prevents the detrimental effects of a maternal high fat diet on the metabolic health in adult offspring, but the effects of maternal exercise on offspring cardiovascular health have not been thoroughly investigated. METHODS To determine the effects of maternal exercise on offspring cardiovascular health, female mice were fed a chow (C; 21% kcal from fat) or high-fat (H; 60% kcal from fat) diet and further subdivided into sedentary (CS, HS) or wheel exercised (CW, HW) prior to pregnancy and throughout gestation. Offspring were maintained in a sedentary state and chow-fed throughout 52 weeks of age and subjected to serial echocardiography and cardiomyocyte isolation for functional and mechanistic studies. RESULTS High-fat fed sedentary dams (HS) produced female offspring with reduced ejection fraction (EF) compared to offspring from chow-fed dams (CS), but EF was preserved in offspring from high-fat fed exercised dams (HW) throughout 52 weeks of age. Cardiomyocytes from HW female offspring had increased kinetics, calcium cycling, and respiration compared to CS and HS offspring. HS offspring had increased oxidation of the RyR2 in cardiomyocytes coupled with increased baseline sarcomere length, resulting in RyR2 overactivity, which was negated in female HW offspring. CONCLUSIONS These data suggest a role for maternal exercise to protect against the detrimental effects of a maternal high-fat diet on female offspring cardiac health. Maternal exercise improved female offspring cardiomyocyte contraction, calcium cycling, respiration, RyR2 oxidation, and RyR2 activity. These data present an important, translatable role for maternal exercise to preserve cardiac health of female offspring and provide insight on mechanisms to prevent the transmission of cardiovascular diseases to subsequent generations.
Collapse
Affiliation(s)
- Kelsey M Pinckard
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Elisa Félix-Soriano
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Surgery, Division of General and Gastrointestinal Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Shanna Hamilton
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Radmila Terentyeva
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Lisa A Baer
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Surgery, Division of General and Gastrointestinal Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Katherine R Wright
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Drew Nassal
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Internal Medicine, Cardiovascular Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Joao Victor Esteves
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Surgery, Division of General and Gastrointestinal Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Eaman Abay
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Vikram K Shettigar
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Mark T Ziolo
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Thomas J Hund
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Internal Medicine, Cardiovascular Medicine, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus, OH, USA
| | - Loren E Wold
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Surgery, Division of Cardiac Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Dmitry Terentyev
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Surgery, Division of General and Gastrointestinal Surgery, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
6
|
Power ML, Muletz-Wolz CR, Bornbusch SL. Microbiome: Mammalian milk microbiomes: sources of diversity, potential functions, and future research directions. REPRODUCTION AND FERTILITY 2024; 5:e230056. [PMID: 38513351 PMCID: PMC11046322 DOI: 10.1530/raf-23-0056] [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: 08/21/2023] [Accepted: 03/18/2024] [Indexed: 03/23/2024] Open
Abstract
Abstract Milk is an ancient, fundamental mammalian adaptation that provides nutrition and biochemical communication to offspring. Microbiomes have been detected in milk of all species studied to date. In this review, we discuss: (a) routes by which microbes may enter milk; (b) evidence for proposed milk microbiome adaptive functions; (c) variation in milk microbiomes across mammals; and (d) future research directions, including suggestions for how to address outstanding questions on the viability and functionality of milk microbiomes. Milk microbes may be sourced from the maternal gastrointestinal tract, oral, skin, and mammary gland microbiomes and from neonatal oral and skin microbiomes. Given the variety of microbial sources, stochastic processes strongly influence milk microbiome assembly, but milk microbiomes appear to be influenced by maternal evolutionary history, diet, environment, and milk nutrients. Milk microbes have been proposed to colonize the neonatal intestinal tract and produce gene and metabolic products that influence physiology, metabolism, and immune system development. Limited epidemiological data indicate that early-life exposure to milk microbes can result in positive, long-term health outcomes. Milk microbiomes can be modified by dietary changes including providing the mother with probiotics and prebiotics. Milk replacers (i.e. infant formula) may benefit from supplementation with probiotics and prebiotics, but data are lacking on probiotics' usefulness, and supplementation should be evidence based. Overall, milk microbiome literature outside of human and model systems is scarce. We highlight the need for mechanistic studies in model species paired with comparative studies across mammals to further our understanding of mammalian milk microbiome evolution. A broader study of milk microbiomes has the potential to inform animal care with relevance to ex situ endangered species. Lay summary Milk is an ancient adaptation that supports the growth and development of mammalian neonates and infants. Beyond its fundamental nutritional function, milk influences all aspects of neonatal development, especially immune function. All kinds of milks so far studied have contained a milk microbiome. In this review, we focus on what is known about the collection of bacterial members found in milk microbiomes. Milk microbiomes include members sourced from maternal and infant microbiomes and they appear to be influenced by maternal evolutionary history, diet, milk nutrients, and environment, as well as by random chance. Once a neonate begins nursing, microbes from milk colonize their gut and produce byproducts that influence their physiology, metabolism, and immune development. Empirical data on milk microbiomes outside of humans and model systems are sparse. Greater study of milk microbiomes across mammals will expand our understanding of mammalian evolution and improve the health of animals under human care.
Collapse
Affiliation(s)
- Michael L Power
- Center for Species Survival, Smithsonian’s National Zoo and Conservation Biology Institute, Washington, District of Columbia, USA
| | - Carly R Muletz-Wolz
- Center for Conservation Genomics, Smithsonian’s National Zoo and Conservation Biology Institute, Washington, District of Columbia, USA
| | - Sally L Bornbusch
- Center for Conservation Genomics, Smithsonian’s National Zoo and Conservation Biology Institute, Washington, District of Columbia, USA
- Department of Nutrition Science, Smithsonian’s National Zoo and Conservation Biology Institute, Washington, District of Columbia, USA
| |
Collapse
|
7
|
Mouzaki M, Woo JG, Divanovic S. Gestational and Developmental Contributors of Pediatric MASLD. Semin Liver Dis 2024; 44:43-53. [PMID: 38423068 DOI: 10.1055/s-0044-1782210] [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] [Indexed: 03/02/2024]
Abstract
Pediatric metabolic dysfunction-associated steatotic liver disease (MASLD) is common and can be seen as early as in utero. A growing body of literature suggests that gestational and early life exposures modify the risk of MASLD development in children. These include maternal risk factors, such as poor cardiometabolic health (e.g., obesity, gestational diabetes, rapid weight gain during pregnancy, and MASLD), as well as periconceptional dietary exposures, degree of physical activity, intestinal microbiome, and smoking. Paternal factors, such as diet and obesity, also appear to play a role. Beyond gestation, early life dietary exposures, as well as the rate of infant weight gain, may further modify the risk of future MASLD development. The mechanisms linking parental health and environmental exposures to pediatric MASLD are complex and not entirely understood. In conclusion, investigating gestational and developmental contributors to MASLD is critical and may identify future interventional targets for disease prevention.
Collapse
Affiliation(s)
- Marialena Mouzaki
- Divisions of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Jessica G Woo
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
8
|
Pereira KHNP, Fuchs KDM, Mendonça JC, Xavier GM, Knupp FC, Lourenço MLG. Topics on maternal, fetal and neonatal immunology of dogs and cats. Vet Immunol Immunopathol 2023; 266:110678. [PMID: 38016336 DOI: 10.1016/j.vetimm.2023.110678] [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: 09/19/2023] [Revised: 11/05/2023] [Accepted: 11/07/2023] [Indexed: 11/30/2023]
Abstract
Birth and the first few weeks of age are critical periods of developing the immune system of puppies and kittens and adapting to an environment containing a variety of infectious agents. The survival rate during these periods depends mainly on the newborn's immune capacity to prevent and combat infections. Although most components of innate and adaptive immunity are present at birth, responses are slow and immature compared to adults. Due to immunological immaturity and the endotheliochorial placental structure, circulating concentrations of immunoglobulins in dogs and cats at birth are quite low. Thus, newborns need a prompt and immediate immune response, which is essentially provided by defense cells and maternal antibodies via colostrum. Failure to ingest colostrum is correlated with high mortality rates in the neonatal period. Concurrently, factors related to pregnant, such as pregnancy physiological immunosuppression and nutritional and health states, can directly influence newborn immunity and health. Therefore, understanding the maternal and neonatal immunological aspects, importance of colostrum, risk factors for failure to transfer passive immunity and colostrum substitute strategies are essential to ensure the survival of the litter.
Collapse
Affiliation(s)
- Keylla Helena Nobre Pacífico Pereira
- Veterinary Neonatology Research Group, Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Kárita da Mata Fuchs
- Veterinary Neonatology Research Group, Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Júlia Cosenza Mendonça
- Veterinary Neonatology Research Group, Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Gleice Mendes Xavier
- Veterinary Neonatology Research Group, Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Fabíola Cardoso Knupp
- Veterinary Neonatology Research Group, Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Maria Lucia Gomes Lourenço
- Veterinary Neonatology Research Group, Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil.
| |
Collapse
|
9
|
Velumula PK, Elbakoush F, Elfadeel H, Lulic-Botica M, Natarajan G, Bajaj M. Comparative Growth Outcomes in Preterm Infants Fed Either Mother's Own Milk or Donor Human Milk. Breastfeed Med 2023; 18:300-306. [PMID: 36971613 DOI: 10.1089/bfm.2022.0224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/19/2023]
Abstract
Objective: To compare growth velocity (GV) in preterm infants fed mother's own milk (MOM) fortified with human milk-based fortifier (HMBF) to those who received donor human milk (DHM) fortified with HMBF. Study Design: A retrospective study of preterm infants with birth weight <1,250 g receiving an exclusive human milk diet. Maternal and infant charts were reviewed for feeding, growth, and short-term neonatal morbidities. Results: On regression analysis, after adjusting (gestational age, multiple births, antenatal steroids, and small for gestational age), no significant difference was observed between the two groups in GV from birth to 32 weeks postmenstrual age (β-coefficient 0.83, 95% confidence interval [CI]: -0.47 to 2.14, p = 0.21), GV from the day of regaining of birth weight to discharge (β-coefficient -0.015, 95% CI: -1.08 to 1.05, p = 0.98). The rate of Grade 3 and 4 intraventricular hemorrhage was significantly higher in the DHM group (19.6% compared to 5.5% in MOM, p = 0.03). Conclusion: At our institution, there was no difference in GV of preterm infants fed HMBF-fortified MOM versus HMBF-fortified DBM.
Collapse
Affiliation(s)
| | - Faesal Elbakoush
- Department of Pediatrics, Children's Hospital of Michigan, Detroit, Michigan, USA
- Department of Pediatrics, Hutzel Women's Hospital/Detroit Medical Center, Detroit, Michigan, USA
| | - Hiba Elfadeel
- Department of Pediatrics, Children's Hospital of Michigan, Detroit, Michigan, USA
- Department of Pediatrics, Hutzel Women's Hospital/Detroit Medical Center, Detroit, Michigan, USA
| | - Mirjana Lulic-Botica
- Department of Pediatrics, Hutzel Women's Hospital/Detroit Medical Center, Detroit, Michigan, USA
- Department of Pharmacy, Wayne State University, Detroit, Michigan, USA
| | - Girija Natarajan
- Department of Pediatrics, Children's Hospital of Michigan, Detroit, Michigan, USA
- Department of Pediatrics, Hutzel Women's Hospital/Detroit Medical Center, Detroit, Michigan, USA
- Department of Pediatrics, Central Michigan University, Detroit, Michigan, USA
| | - Monika Bajaj
- Department of Pediatrics, Children's Hospital of Michigan, Detroit, Michigan, USA
- Department of Pediatrics, Hutzel Women's Hospital/Detroit Medical Center, Detroit, Michigan, USA
- Department of Pediatrics, Central Michigan University, Detroit, Michigan, USA
| |
Collapse
|
10
|
Effect of an exclusive human milk diet on feeding tolerance in preterm infants. J Perinatol 2022; 42:1070-1075. [PMID: 35184144 DOI: 10.1038/s41372-022-01348-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 01/13/2022] [Accepted: 02/03/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To compare the time to full enteral feeds in preterm infants fed exclusive human milk (EHM) - mother's own milk (MOM) fortified with human milk-based fortifier (HMBF), to those who received partial human milk (PHM) - MOM fortified with bovine milk-based fortifier (BMBF), and exclusive formula. STUDY DESIGN A single-center retrospective study of infants with birth weight <1250 g from 2013 to 2018. Data on feeding, growth and other short-term neonatal morbidities were collected. RESULTS On regression analysis, time to full enteral feeds was significantly higher in PHM compared to EHM group (β-coefficient 4.14, 95% CI 0.00-8.29) and formula-fed group compared to EHM (β-coefficient 4.3, 95% CI 0.32-8.20). No significant differences in growth velocity, length of stay and other morbidities were found between the groups. CONCLUSION Infants in EHM had better feeding tolerance and reached their enteral feed goals sooner compared to PHM and formula-fed groups.
Collapse
|
11
|
Calvo-Lerma J, Bueno-Llamoga P, Bäuerl C, Cortés-Macias E, Selma-Royo M, Pérez-Cano F, Lerin C, Martínez-Costa C, Collado MC. Persistence of Anti SARS-CoV-2 Antibodies in Breast Milk from Infected and Vaccinated Women after In Vitro-Simulated Gastrointestinal Digestion. Nutrients 2022; 14:2117. [PMID: 35631258 PMCID: PMC9147794 DOI: 10.3390/nu14102117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/09/2022] [Accepted: 05/14/2022] [Indexed: 02/04/2023] Open
Abstract
Breastfeeding is key for infant development and growth. Breast milk contains different bioactive compounds including antibodies. Recent studies have demonstrated the presence of breast milk SARS-CoV-2 antibodies after maternal infection and vaccination. However, the potential impact on the infant has not been explored yet. As a first step, we aimed at assessing the potential persistence of SARS-CoV-2 IgA and IgG antibodies from infected and vaccinated women in the gastrointestinal tract of the infants by means of an in vitro-simulated gastrointestinal digestion approach. Breast milk samples from 10 lactating women receiving mRNA vaccination against SARS-CoV-2 (n = 5 with BNT162b2 mRNA and n = 5 with mRNA-1273) and also, COVID-19 infected (n = 5) were included. A control group with women with no exposure to the virus (n = 10 pre-pandemic) were also studied. The presence of IgA and IgG SARS-CoV-2 antibody levels was determined by ELISA after the gastric and intestinal stages. The impact of digested antibodies on infant gut microbiota was tested by simulating colonic fermentation with two different fecal inoculums: infants from vaccinated and non-vaccinated mothers. Specific gut microbial groups were tested by targeted qPCR. In vitro infant gastrointestinal digestion significantly decreased the levels of both anti-SARS-CoV-2 IgA and IgG. However, both remained resistant in all the study groups except in that evaluating breast milk samples from infected women, in which IgG was degraded below the cut-off values in the intestinal phase. No effect of the antibodies on microbiota were identified after digestion. In conclusion, antibody levels against SARS-CoV-2 are reduced after in vitro-simulated gastrointestinal tract but remain present, so a positive biological effect could be expected from this infant immunization pathway.
Collapse
Affiliation(s)
- Joaquim Calvo-Lerma
- Institute of Agrochemistry and Food Technology—National Research Council (IATA-CSIC), 46980 Paterna, Valencia, Spain; (P.B.-L.); (C.B.); (E.C.-M.); (M.S.-R.)
| | - Pierre Bueno-Llamoga
- Institute of Agrochemistry and Food Technology—National Research Council (IATA-CSIC), 46980 Paterna, Valencia, Spain; (P.B.-L.); (C.B.); (E.C.-M.); (M.S.-R.)
| | - Christine Bäuerl
- Institute of Agrochemistry and Food Technology—National Research Council (IATA-CSIC), 46980 Paterna, Valencia, Spain; (P.B.-L.); (C.B.); (E.C.-M.); (M.S.-R.)
| | - Erika Cortés-Macias
- Institute of Agrochemistry and Food Technology—National Research Council (IATA-CSIC), 46980 Paterna, Valencia, Spain; (P.B.-L.); (C.B.); (E.C.-M.); (M.S.-R.)
| | - Marta Selma-Royo
- Institute of Agrochemistry and Food Technology—National Research Council (IATA-CSIC), 46980 Paterna, Valencia, Spain; (P.B.-L.); (C.B.); (E.C.-M.); (M.S.-R.)
| | - Francisco Pérez-Cano
- Section of Physiology, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Catalonia, Spain;
- Institute of Research in Nutrition and Food Safety (INSA), University of Barcelona (UB), 08921 Santa Coloma de Gramenet, Catalonia, Spain
| | - Carles Lerin
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, 08950 Barcelona, Catalonia, Spain;
| | - Cecilia Martínez-Costa
- Department of Pediatrics, Hospital Clínico Universitario, University of Valencia, 46010 Valencia, Valencia, Spain;
- Nutrition Research Group of INCLIVA, 46010 Valencia, Valencia, Spain
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology—National Research Council (IATA-CSIC), 46980 Paterna, Valencia, Spain; (P.B.-L.); (C.B.); (E.C.-M.); (M.S.-R.)
| |
Collapse
|
12
|
de Weerth C, Aatsinki AK, Azad MB, Bartol FF, Bode L, Collado MC, Dettmer AM, Field CJ, Guilfoyle M, Hinde K, Korosi A, Lustermans H, Mohd Shukri NH, Moore SE, Pundir S, Rodriguez JM, Slupsky CM, Turner S, van Goudoever JB, Ziomkiewicz A, Beijers R. Human milk: From complex tailored nutrition to bioactive impact on child cognition and behavior. Crit Rev Food Sci Nutr 2022; 63:7945-7982. [PMID: 35352583 DOI: 10.1080/10408398.2022.2053058] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human milk is a highly complex liquid food tailor-made to match an infant's needs. Beyond documented positive effects of breastfeeding on infant and maternal health, there is increasing evidence that milk constituents also impact child neurodevelopment. Non-nutrient milk bioactives would contribute to the (long-term) development of child cognition and behavior, a process termed 'Lactocrine Programming'. In this review we discuss the current state of the field on human milk composition and its links with child cognitive and behavioral development. To promote state-of-the-art methodologies and designs that facilitate data pooling and meta-analytic endeavors, we present detailed recommendations and best practices for future studies. Finally, we determine important scientific gaps that need to be filled to advance the field, and discuss innovative directions for future research. Unveiling the mechanisms underlying the links between human milk and child cognition and behavior will deepen our understanding of the broad functions of this complex liquid food, as well as provide necessary information for designing future interventions.
Collapse
Affiliation(s)
- Carolina de Weerth
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, EN Nijmegen, The Netherlands
| | - Anna-Katariina Aatsinki
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Meghan B Azad
- Department of Pediatrics and Child Health, Manitoba Interdisciplinary Lactation Centre, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Frank F Bartol
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Lars Bode
- Department of Pediatrics and Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California San Diego, La Jolla, California, USA
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Amanda M Dettmer
- Yale Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, College of Basic and Applied Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Meagan Guilfoyle
- Department of Anthropology, Indiana University, Bloomington, Indiana, USA
| | - Katie Hinde
- School of Human Evolution and Social Change, Arizona State University, Tempe, Arizona, USA
| | - Aniko Korosi
- Swammerdam Institute for Life Sciences, Center for Neuroscience, Brain Plasticity group, University of Amsterdam, Amsterdam, The Netherlands
| | - Hellen Lustermans
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, EN Nijmegen, The Netherlands
| | - Nurul Husna Mohd Shukri
- Department of Nutrition, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Sophie E Moore
- Department of Women & Children's Health, King's College London, St Thomas' Hospital, London, UK
- School of Hygiene and Tropical Medicine, Nutrition Theme, MRC Unit The Gambia and the London, Fajara, The GambiaBanjul
| | - Shikha Pundir
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Juan Miguel Rodriguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Carolyn M Slupsky
- Department of Nutrition and Department of Food Science and Technology, University of California, Davis, California, USA
| | - Sarah Turner
- Department of Community Health Sciences, Manitoba Interdisciplinary Lactation Centre, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Johannes B van Goudoever
- Department of Pediatrics, Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Amsterdam, The Netherlands
| | - Anna Ziomkiewicz
- Department of Anthropology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Roseriet Beijers
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, EN Nijmegen, The Netherlands
- Department of Social Development, Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
13
|
Taylor SN, Fenton TR, Groh-Wargo S, Gura K, Martin CR, Griffin IJ, Rozga M, Moloney L. Exclusive Maternal Milk Compared With Exclusive Formula on Growth and Health Outcomes in Very-Low-Birthweight Preterm Infants: Phase II of the Pre-B Project and an Evidence Analysis Center Systematic Review. Front Pediatr 2022; 9:793311. [PMID: 35280446 PMCID: PMC8913886 DOI: 10.3389/fped.2021.793311] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/27/2021] [Indexed: 11/13/2022] Open
Abstract
As part of the Pre-B Project, a systematic review was conducted to evaluate associations between exclusive maternal milk (≥75%) intake and exclusive formula intake and growth and health outcomes in very-low-birthweight (VLBW) preterm infants. The protocols from the Academy of Nutrition and Dietetics' Evidence Analysis Center and the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) checklist were followed. Thirteen observational studies were included; 11 studies reported data that could be synthesized in a pooled analysis. The evidence is very uncertain (very low quality) about the effect of exclusive maternal milk on all outcomes due to observational study designs and risk of selection, performance, detection, and reporting bias in most of the included studies. Very-low-quality evidence suggested that providing VLBW preterm infants with exclusive maternal milk was not associated with mortality, risk of necrotizing enterocolitis, sepsis, or developing bronchopulmonary dysplasia, as compared with exclusive preterm formula, but exclusive maternal milk was associated with a lower risk of retinopathy of prematurity (very low certainty). Results may change when additional studies are conducted. There was no difference in weight, length, and head circumference gain between infants fed fortified exclusive maternal milk and infants receiving exclusive preterm formula; however, weight and length gain were lower in infants fed non-fortified exclusive maternal milk. Given the observational nature of human milk research, cause-and-effect evidence was lacking for VLBW preterm infants. Systematic Review Registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=86829, PROSPERO ID: CRD42018086829.
Collapse
Affiliation(s)
- Sarah N. Taylor
- Division of Neonatology, Department of Pediatrics, Yale School of Medicine, New Haven, CT, United States
| | - Tanis R. Fenton
- Community Health Sciences, Institute of Public Health, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Nutrition Services, Alberta Health Services, Calgary, AB, Canada
- Nutrition Services, Alberta Health Services, Calgary, AB, Canada
| | - Sharon Groh-Wargo
- Departments of Nutrition and Pediatrics, Case Western Reserve University at MetroHealth Medical Center, Cleveland, OH, United States
| | - Kathleen Gura
- Clinical Research Program, Department of Pharmacy, Boston Children's Hospital, Boston, MA, United States
| | - Camilia R. Martin
- Division of Translational Research, Department of Neonatology, Harvard Medical School, Neonatal Intensive Care Unit (NICU), Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Ian J. Griffin
- Biomedical Research Institute of New Jersey, Cedar Knolls, NJ, United States
- Department of Pediatrics, Morristown Medical Center, Morristown, NJ, United States
| | - Mary Rozga
- Academy of Nutrition and Dietetics, Evidence Analysis Center, Chicago, IL, United States
| | - Lisa Moloney
- Academy of Nutrition and Dietetics, Evidence Analysis Center, Chicago, IL, United States
| |
Collapse
|
14
|
Vacca M, Raspini B, Calabrese FM, Porri D, De Giuseppe R, Chieppa M, Liso M, Cerbo RM, Civardi E, Garofoli F, Cena H, De Angelis M. The establishment of the gut microbiota in 1-year-aged infants: from birth to family food. Eur J Nutr 2022; 61:2517-2530. [PMID: 35211851 PMCID: PMC9279275 DOI: 10.1007/s00394-022-02822-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/25/2022] [Indexed: 12/14/2022]
Abstract
Purpose With the aim of characterizing the gastrointestinal (GI) microbiota and contextually determine how different prenatal, perinatal, and postnatal factors affected its composition in early childhood, infants were enrolled in a longitudinal-prospective study named “A.MA.MI.” (Alimentazione MAmma e bambino nei primi MIlle giorni; NCT04122612, October 2019). Methods Forty-five fecal samples were collected at 12 months of infants’ age, identified as the 3rd follow-up (T3). The evaluated variables were pre-gestational weight and weight gain during pregnancy, delivery mode, feeding, timing of weaning, and presence/absence of older siblings. Fecal alpha and beta-diversities were analyzed. Noteworthy, to determine the impact of the influencing factors, multivariate analyses were conducted. Results At T3, all prenatal and perinatal variables did not result to be significant whereas, among the postnatal variables, type of milk-feeding and weaning showed the greatest contribution in shaping the microbiota. Although aged 1 year, infants exclusively breastfed until 6 months were mainly colonized by Lactobacillaceae and Enterobacteriaceae. Differently, Bacteroidaceae characterized the microbiota of infants that were never breastfed in an exclusive way. Moreover, although an early introduction of solid foods determined higher values of Faith’s PD, high abundances of Ruminococcaceae and Faecalibacterium mainly associated with infants weaned after the 4th month of age. Conclusion The microbial colonization during the first year of life is likely affected by a simultaneous effect of multiple variables playing a significant role at different times. Therefore, these data contribute to add evidence concerning the complex multifactorial interaction between GI microbiota and various stimuli affecting infants during the early stages of life. Supplementary Information The online version contains supplementary material available at 10.1007/s00394-022-02822-1.
Collapse
Affiliation(s)
- Mirco Vacca
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
| | - Benedetta Raspini
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | | | - Debora Porri
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Rachele De Giuseppe
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Marcello Chieppa
- Institute of Research, National Institute of Gastroenterology "S. de Bellis", Castellana Grotte, Italy
| | - Marina Liso
- Institute of Research, National Institute of Gastroenterology "S. de Bellis", Castellana Grotte, Italy
| | - Rosa Maria Cerbo
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elisa Civardi
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Francesca Garofoli
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Hellas Cena
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy. .,Unit of Internal Medicine and Endocrinology, Clinical Nutrition and Dietetics Service, ICS Maugeri IRCCS, Pavia, Italy.
| | - Maria De Angelis
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
15
|
Liang N, Beverly RL, Scottoline BP, Dallas DC. Peptides Derived from In Vitro and In Vivo Digestion of Human Milk Are Immunomodulatory in THP-1 Human Macrophages. J Nutr 2021; 152:331-342. [PMID: 34601601 PMCID: PMC8754566 DOI: 10.1093/jn/nxab350] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/11/2021] [Accepted: 09/23/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Milk proteins contain many encrypted bioactive peptides. Whether these bioactive peptides are released in the infant intestine and exert immunomodulatory activity remains unknown. OBJECTIVE This study examined in vitro immunomodulatory activities of peptides from in vitro- and in vivo-digested human milk. METHODS Peptides were extracted from in vitro-digested human milk and pooled intestinal samples from 8 infants fed human milk. Peptides extracted from in vitro-digested samples were fractionated. The in vitro effects of these peptides and fractions on the secretion of TNF-α and IL-8 in LPS-treated human immune THP-1 macrophages were evaluated. The significance of differences between in vitro peptide fraction treatment and control on cytokine production was analyzed by t test. LC-MS/MS-based peptidomics was conducted to identify the peptides. The peptides were screened for potential bioactivity using a sequence homology search using the Milk Bioactive Peptide Database (MBPDB). RESULTS Six fractions of the peptide mixture extracted from the in vitro-digested human milk significantly inhibited TNF-α production by LPS-challenged THP-1 macrophages. Fractions F4, F8, F11, F14, and F17 attenuated IL-8 secretion, and F6/7 and F18 increased IL-8 secretion. Peptides extracted from the pooled in vivo intestinal samples attenuated both TNF-α and IL-8 secretion. There were 266 and 418 peptides identified in the in vitro and in vivo samples, respectively. Among the peptides, 34 and 50 in the in vitro and in vivo samples, respectively, had >80% sequence similarity to bioactive peptides in the MBPDB. CONCLUSIONS Peptides released by in vitro and in vivo infant digestion of human milk were immunomodulatory in human immune cells; fractions F4, F8, and F11 were anti-inflammatory; and F6/7 and F18 were proinflammatory. Thirteen peptides were present in all fractions with anti-inflammatory activity, and 38 peptides were present in all fractions with proinflammatory activity. These peptides potentially contributed to the observed immunomodulatory activity of the peptide mixtures.
Collapse
Affiliation(s)
- Ningjian Liang
- Nutrition Program, School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA
| | - Robert L Beverly
- Nutrition Program, School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA
| | - Brian P Scottoline
- Department of Pediatrics, Oregon Health and Science University, Portland, OR, USA
| | | |
Collapse
|
16
|
Koosha RZ, Fazel P, Sedighian H, Behzadi E, Ch MH, Imani Fooladi AA. The impact of the gut microbiome on toxigenic bacteria. Microb Pathog 2021; 160:105188. [PMID: 34530074 DOI: 10.1016/j.micpath.2021.105188] [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: 04/18/2021] [Revised: 07/05/2021] [Accepted: 09/09/2021] [Indexed: 10/20/2022]
Abstract
Millions of symbiotic and pathogenic microorganisms known as microbiota colonize the host body. The microbiome plays an important role in human health and colonizes hundreds of different species of multicellular organisms so that they are introduced as the metaorganisms. Changes in the microbial population of the gut microbiome may cause resistance to pathogenic bacteria-induced infection. Understanding the principles of Host-Microbiota Interactions (HMIs) is important because it clarifies our insight towards the mechanisms of infections established in the host. Interactions between the host and the microbiota help answer the question of how a microorganism can contribute to the health or disease of the host. Microbiota can increase host resistance to colonization of pathogenic species. Studying the HMIs network can in several ways delineate the pathogenic mechanisms of pathogens and thereby help to increase useful and novel therapeutic pathways. For example, the potentially unique microbial effects that target the distinct host or interfere with the endogenous host interactions can be identified. In addition, the way mutations in essential proteins in the host and/or in the microbes can influence the interactions between them may be determined. Furthermore, HMIs help in identifying host cell regulatory modules.
Collapse
Affiliation(s)
- Roohollah Zarei Koosha
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Parvindokht Fazel
- Department of Microbiology, Fars Science and Research Branch, Islamic Azad University, Fars, Iran; Department of Microbiology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Hamid Sedighian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Elham Behzadi
- Department of Microbiology, College of Basic Sciences, Shahr-e-Qods Branch, Islamic Azad University, Tehran, Iran
| | - Mojtaba Hedayati Ch
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Microbial Toxins Physiology Group, Universal Scientific Education and Research Network, Rasht, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Ley D, Beghin L, Morcel J, Flamein F, Garabedian C, Accart B, Drumez E, Labreuche J, Gottrand F, Hermann E. Impact of early life nutrition on gut health in children: a prospective clinical study. BMJ Open 2021; 11:e050432. [PMID: 34489289 PMCID: PMC8422494 DOI: 10.1136/bmjopen-2021-050432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 08/03/2021] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION The first 1000 days of life could contribute to individual susceptibility to the later development of chronic non-communicable diseases. Nutrition in early life appears to be an important determinant factor for a sustainable child's health. In this study, we propose to investigate the impact of exclusive breast feeding on gut health in children. METHODS AND ANALYSIS A prospective cohort of newborns (n=350) will be recruited at birth and followed up to 4 years of age. The main objective is to evaluate the link between exclusive breast feeding for at least 3 months and the gut health of the child at 4 years. The primary endpoint of assessment of gut health will be based on the non-invasive measurement of faecal secretory IgA (sIgA) as a sensitive biomarker of the intestinal ecosystem. The presence of gastrointestinal disorders will be defined according to the clinical criteria of Rome IV. Information on parent's nutritional habits and life style, breastfeeding duration and child's complementary feeding will be collected along the follow-up. Cord blood cells and plasma at birth will be purified for further analysis. The meconium and stools collected at birth, 6 months, 2 years and 4 years of age will allow sIgA analysis. ETHICS AND DISSEMINATION This clinical study has obtained the approval from the national ethical committee. We plan to publish the results of the study in peer-review journals and by means of national and international conference. TRIAL REGISTRATION NUMBER NCT04195425.
Collapse
Affiliation(s)
- Delphine Ley
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Laurent Beghin
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Jules Morcel
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Florence Flamein
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Charles Garabedian
- Univ. Lille, CHU Lille, Department of Obstetrics & Gynecology, F-59000 Lille, France
- Univ. Lille, CHU Lille, ULR 2694-METRICS: évaluation des technologies de santé et des pratiques médicales, F-59000 Lille, France
| | | | - Elodie Drumez
- Univ. Lille, CHU Lille, ULR 2694-METRICS: évaluation des technologies de santé et des pratiques médicales, F-59000 Lille, France
- CHU Lille, Department of Biostatistics, F-59000 Lille, France
| | - Julien Labreuche
- Univ. Lille, CHU Lille, ULR 2694-METRICS: évaluation des technologies de santé et des pratiques médicales, F-59000 Lille, France
- CHU Lille, Department of Biostatistics, F-59000 Lille, France
| | - Frederic Gottrand
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Emmanuel Hermann
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| |
Collapse
|
18
|
Han G, Nishigawa T, Ikeda H, Hamada M, Yang H, Maesono S, Aso K, Jing A, Furuse M, Zhang R. Dysregulated metabolism and behaviors by disrupting gut microbiota in prenatal and neonatal mice. Anim Sci J 2021; 92:e13566. [PMID: 34170061 DOI: 10.1111/asj.13566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/28/2021] [Accepted: 05/07/2021] [Indexed: 11/26/2022]
Abstract
The live microbiota ecosystem in the intestine plays a critical role in maintaining the normal physiological and psychological functions in both animals and human beings. However, the chronic effect of microbiota disturbances during prenatal and neonatal developing periods on animal's health remains less studied. In the current study, pregnant ICR mice were fed with an antibiotic diet (7-g nebacitin [bacitracin-neomycin sulphate 2:1]/kg standard diet) from day 14 of conception, and their offspring were provided with the same diet till the termination of the experiments. Dams treated with antibiotics showed increased body weight along with enlarged gut. Antibiotic-treated offspring revealed decreased bodyweight, increased food, water, and sucrose intake. Administration of antibiotics affected corticosterone responsivity to acute 20 min restraint challenge in male pups. In behavior tests, female pups showed decreased movement in open field while male pups revealed decreased latency to open arms in elevated plus maze test and immobility time in tail suspension test. Together, these results suggested that early antibiotic exposure may impact on the food intake, body weight gain, and emotional behavior regulation in mice.
Collapse
Affiliation(s)
- Guofeng Han
- Laboratory of Regulation in Metabolism and Behavior, Department of Bioresource Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan.,Department of Animal Nutrition and Food Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Takuma Nishigawa
- Laboratory of Regulation in Metabolism and Behavior, Department of Bioresource Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Hiromi Ikeda
- Laboratory of Regulation in Metabolism and Behavior, Department of Bioresource Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Mizuki Hamada
- Laboratory of Regulation in Metabolism and Behavior, Department of Bioresource Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Hui Yang
- Laboratory of Regulation in Metabolism and Behavior, Department of Bioresource Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Saori Maesono
- Laboratory of Regulation in Metabolism and Behavior, Department of Bioresource Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Kenta Aso
- Laboratory of Regulation in Metabolism and Behavior, Department of Bioresource Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Ashley Jing
- Laboratory of Regulation in Metabolism and Behavior, Department of Bioresource Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Mitsuhiro Furuse
- Laboratory of Regulation in Metabolism and Behavior, Department of Bioresource Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Rong Zhang
- Laboratory of Regulation in Metabolism and Behavior, Department of Bioresource Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan.,Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,School of Medicine, Northwest University, Xi'An, China
| |
Collapse
|
19
|
Torun A, Hupalowska A, Trzonkowski P, Kierkus J, Pyrzynska B. Intestinal Microbiota in Common Chronic Inflammatory Disorders Affecting Children. Front Immunol 2021; 12:642166. [PMID: 34163468 PMCID: PMC8215716 DOI: 10.3389/fimmu.2021.642166] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
The incidence and prevalence rate of chronic inflammatory disorders is on the rise in the pediatric population. Recent research indicates the crucial role of interactions between the altered intestinal microbiome and the immune system in the pathogenesis of several chronic inflammatory disorders in children, such as inflammatory bowel disease (IBD) and autoimmune diseases, such as type 1 diabetes mellitus (T1DM) and celiac disease (CeD). Here, we review recent knowledge concerning the pathogenic mechanisms underlying these disorders, and summarize the facts suggesting that the initiation and progression of IBD, T1DM, and CeD can be partially attributed to disturbances in the patterns of composition and abundance of the gut microbiota. The standard available therapies for chronic inflammatory disorders in children largely aim to treat symptoms. Although constant efforts are being made to maximize the quality of life for children in the long-term, sustained improvements are still difficult to achieve. Additional challenges are the changing physiology associated with growth and development of children, a population that is particularly susceptible to medication-related adverse effects. In this review, we explore new promising therapeutic approaches aimed at modulation of either gut microbiota or the activity of the immune system to induce a long-lasting remission of chronic inflammatory disorders. Recent preclinical studies and clinical trials have evaluated new approaches, for instance the adoptive transfer of immune cells, with genetically engineered regulatory T cells expressing antigen-specific chimeric antigen receptors. These approaches have revolutionized cancer treatments and have the potential for the protection of high-risk children from developing autoimmune diseases and effective management of inflammatory disorders. The review also focuses on the findings of studies that indicate that the responses to a variety of immunotherapies can be enhanced by strategic manipulation of gut microbiota, thus emphasizing on the importance of proper interaction between the gut microbiota and immune system for sustained health benefits and improvement of the quality of life of pediatric patients.
Collapse
Affiliation(s)
- Anna Torun
- Chair and Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland
| | - Anna Hupalowska
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdansk, Gdansk, Poland
| | - Jaroslaw Kierkus
- Department of Gastroenterology, Hepatology, Feeding Disorders and Pediatrics, The Children's Memorial Health Institute, Warsaw, Poland
| | - Beata Pyrzynska
- Chair and Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
20
|
Maternal Distress and Social Support Are Linked to Human Milk Immune Properties. Nutrients 2021; 13:nu13061857. [PMID: 34072410 PMCID: PMC8226629 DOI: 10.3390/nu13061857] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 12/31/2022] Open
Abstract
Possible alterations of maternal immune function due to psychological stress may reflect immunoactive factor levels in breast milk. This study aimed to assess the association between maternal distress and breast milk levels of secretory IgA (SIgA), IgM, IgG, and lactoferrin (LF). We hypothesized that this association is moderated by maternal social support achieved from others during lactation. The study group included 103 lactating mothers and their healthy five-month-old infants. Maternal distress was determined based on the State Anxiety Inventory and the level of salivary cortisol. Social support was assessed using the Berlin Social Support Scales. Breast milk samples were collected to test for SIgA, IgM, IgG, and LF using the ELISA method. Milk immunoactive factors were regressed against maternal anxiety, social support, salivary cortisol, and infant gestational age using the general regression model. Maternal anxiety was negatively associated with milk levels of LF (β = -0.23, p = 0.028) and SIgA (β = -0.30, p = 0.004), while social support was positively associated with milk IgG (β = 0.25, p = 0.017). Neither anxiety nor social support were related to milk IgM. No association was found between the level of maternal salivary cortisol and immunoactive factors in milk. Our results suggest that maternal psychological wellbeing and social support may affect milk immune properties.
Collapse
|
21
|
Lactoferrin and Immunoglobulin Concentrations in Milk of Gestational Diabetic Mothers. Nutrients 2021; 13:nu13030818. [PMID: 33801292 PMCID: PMC7998843 DOI: 10.3390/nu13030818] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 01/03/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is associated with an increased risk of having a high-care newborn and has an impact on maternal wellbeing. This study aimed to assess the effect of GDM on the lactoferrin (LF), secretory immunoglobulin A (SIgA), immunoglobulin G (IgG), and immunoglobulin M (IgM) concentrations in early colostrum, colostrum, and transitional milk samples of hyperglycemic (n = 53) and normoglycemic (n = 49) mothers using enzyme-linked immunosorbent assay (ELISA). The concentrations of milk lactoferrin and SIgA, but not IgG and IgM, from hyperglycemic and normoglycemic mothers, showed a similar negative correlation with lactation from the first to the fifteenth day. Apart from early colostral IgG, there were no differences in concentrations of LF and immunoglobulins in milk from hyperglycemic and normoglycemic mothers. For hyperglycemia compensated by diet (GDM G1) or insulin treatment (GDM G2), slight differences were seen for LF and IgG, but not for SIgA and IgM, during an early stage of lactation only. Early colostral IgG and colostral LF of insulin-treated mothers were higher (10.01 ± 4.48 mg/L and 11.50 ± 0.58 g/L, respectively) than for diet-control diabetic mothers (7.65 ± 5.67 mg/L and 8.05 ± 1.38 g/L, respectively). GDM of mothers does not have a significant impact on immunological quality of early milk.
Collapse
|
22
|
Wei H, Wang JY. Role of Polymeric Immunoglobulin Receptor in IgA and IgM Transcytosis. Int J Mol Sci 2021; 22:ijms22052284. [PMID: 33668983 PMCID: PMC7956327 DOI: 10.3390/ijms22052284] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Transcytosis of polymeric IgA and IgM from the basolateral surface to the apical side of the epithelium and subsequent secretion into mucosal fluids are mediated by the polymeric immunoglobulin receptor (pIgR). Secreted IgA and IgM have vital roles in mucosal immunity in response to pathogenic infections. Binding and recognition of polymeric IgA and IgM by pIgR require the joining chain (J chain), a small protein essential in the formation and stabilization of polymeric Ig structures. Recent studies have identified marginal zone B and B1 cell-specific protein (MZB1) as a novel regulator of polymeric IgA and IgM formation. MZB1 might facilitate IgA and IgM transcytosis by promoting the binding of J chain to Ig. In this review, we discuss the roles of pIgR in transcytosis of IgA and IgM, the roles of J chain in the formation of polymeric IgA and IgM and recognition by pIgR, and focus particularly on recent progress in understanding the roles of MZB1, a molecular chaperone protein.
Collapse
Affiliation(s)
- Hao Wei
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China;
| | - Ji-Yang Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China;
- Department of Clinical Immunology, Children’s Hospital of Fudan University, Shanghai 201102, China
- Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
- Correspondence: ; Tel.: +86-(21)-54237957
| |
Collapse
|
23
|
Qi C, Ding M, Li S, Zhou Q, Li D, Yu R, Sun J. Sex-dependent modulation of immune development in mice by secretory IgA-coated Lactobacillus reuteri isolated from breast milk. J Dairy Sci 2021; 104:3863-3875. [PMID: 33612242 DOI: 10.3168/jds.2020-19437] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022]
Abstract
Lactobacilli, commonly present in human breast milk, appear to colonize the neonatal gut and provide protection to infants against various infections, thereby promoting immune development. This study examined the potential probiotic role of breast milk-derived Lactobacillus reuteri FN041 in immune development in mice. The FN041 were gavaged either to BALB/c dams (n = 6/group) during the lactation period or to their offspring (n = 6/sex per intervention) after weaning separately (cointervention). All interventions induced increased intestinal barriers in 5-wk-old offspring, especially in the females. Immunoglobulin A plasmocytes in ileal tissue and secretory IgA (sIgA) in ileal contents increased in all 5-wk-old offspring of cointervention. The activation of mRNA expression of 17 genes was sex-dependent, especially in 5-wk-old offspring. Broader genes were regulated in female mice. The effect of cointervention on the Shannon index of total microbiota is sex-related. The Shannon index of sIgA-coated microbiota increased in both sexes. The sIgA-coated microbiota showed intergroup differences according to β diversity, especially in female mice that showed an increase in Bifidobacterium of Actinobacteria. The sIgA-coated Bifidobacterium was positively correlated with mRNA expression of Tlr9. The sIgA-coated Lactobacillus in male offspring was negatively correlated with mRNA expression of Cldn2. In conclusion, L. reuteri FN041 promoted the production of intestinal sIgA and the expression of genes related to antimicrobial peptides in the offspring and enhanced the function of the mucosal barrier, depending on sex and treatment manner.
Collapse
Affiliation(s)
- Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China
| | - Mengfan Ding
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China
| | - Shuangqi Li
- Guangzhou Fine Nutrition Research Center, Guangzhou, 510700, PR China
| | - Qin Zhou
- Department of Neonatology, The Affiliated Wuxi Maturity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, PR China
| | - Duo Li
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China
| | - Renqiang Yu
- Department of Neonatology, The Affiliated Wuxi Maturity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, PR China.
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China.
| |
Collapse
|
24
|
Cucinotta U, Romano C, Dipasquale V. Diet and Nutrition in Pediatric Inflammatory Bowel Diseases. Nutrients 2021; 13:655. [PMID: 33671453 PMCID: PMC7922138 DOI: 10.3390/nu13020655] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/10/2021] [Accepted: 02/13/2021] [Indexed: 12/17/2022] Open
Abstract
Both genetic and environmental factors are involved in the onset of inflammatory bowel disease (IBD). In particular, diet composition is suspected to significantly contribute to IBD risk. In recent years, major interest has raised about the role of nutrition in disease pathogenesis and course, and many studies have shown a clear link between diet composition and intestinal permeability impairment. Moreover, many IBD-related factors, such as poor dietary intake, nutrients loss and drugs interact with nutritional status, thus paving the way for the development of many therapeutic strategies in which nutrition represents the cornerstone, either as first-line therapy or as reversing nutritional deficiencies and malnutrition in IBD patients. Exclusive enteral nutrition (EEN) is the most rigorously supported dietary intervention for the treatment of Crohn's Disease (CD), but is burdened by a low tolerability, especially in pediatric patients. Promising alternative regimens are represented by Crohn's Disease Exclusion Diet (CDED), and other elimination diets, whose use is gradually spreading. The aim of the current paper is to provide a comprehensive and updated overview on the latest evidence about the role of nutrition and diet in pediatric IBD, focusing on the different nutritional interventions available for the management of the disease.
Collapse
Affiliation(s)
| | - Claudio Romano
- Pediatric Gastroenterology and Cystic Fibrosis Unit, Department of Human Pathology in Adulthood and Childhood “G. Barresi”, University of Messina, 98124 Messina, Italy; (U.C.); (V.D.)
| | | |
Collapse
|
25
|
The Gut‒Breast Axis: Programming Health for Life. Nutrients 2021; 13:nu13020606. [PMID: 33673254 PMCID: PMC7917897 DOI: 10.3390/nu13020606] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/03/2021] [Accepted: 02/09/2021] [Indexed: 12/16/2022] Open
Abstract
The gut is a pivotal organ in health and disease. The events that take place in the gut during early life contribute to the programming, shaping and tuning of distant organs, having lifelong consequences. In this context, the maternal gut plays a quintessence in programming the mammary gland to face the nutritional, microbiological, immunological, and neuroendocrine requirements of the growing infant. Subsequently, human colostrum and milk provides the infant with an impressive array of nutrients and bioactive components, including microbes, immune cells, and stem cells. Therefore, the axis linking the maternal gut, the breast, and the infant gut seems crucial for a correct infant growth and development. The aim of this article is not to perform a systematic review of the human milk components but to provide an insight of their extremely complex interactions, which render human milk a unique functional food and explain why this biological fluid still truly remains as a scientific enigma.
Collapse
|
26
|
de Castro MM, Pascoal LB, Steigleder KM, Siqueira BP, Corona LP, Ayrizono MDLS, Milanski M, Leal RF. Role of diet and nutrition in inflammatory bowel disease. World J Exp Med 2021; 11:1-16. [PMID: 33585174 PMCID: PMC7852575 DOI: 10.5493/wjem.v11.i1.1] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/02/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel diseases (IBDs) are closely linked to nutrition. The latest research indicates that diet and nutrition are significantly involved in the etiopathogenesis of the disease, although their specific role throughout its clinical course still remains unclear. This study reviewed how diet and nutrition are associated with IBD development and management. Even though specific diets have been shown to bring about positive outcomes, there is currently no scientific consensus regarding an appropriate diet that would benefit all IBD patients. We suggest that individualized dietary recommendations are of the greatest importance and that diets should be planned to provide individual IBD patients with specific nutrient requirements while keeping all the clinical aspects of the patients in mind. Further research is clearly necessary to investigate nutritional factors involved in IBD development and, especially, to evaluate the applications of the diets during the course of the disease.
Collapse
Affiliation(s)
- Marina Moreira de Castro
- IBD Research Laboratory, Colorectal Surgery Unit, School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-878, São Paulo, Brazil
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas (UNICAMP), Campinas 13083-878, São Paulo, Brazil
| | - Lívia Bitencourt Pascoal
- IBD Research Laboratory, Colorectal Surgery Unit, School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-878, São Paulo, Brazil
| | - Karine Mariane Steigleder
- IBD Research Laboratory, Colorectal Surgery Unit, School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-878, São Paulo, Brazil
| | - Beatriz Piatezzi Siqueira
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas (UNICAMP), Limeira 13484-350, São Paulo, Brazil
| | - Ligiana Pires Corona
- Laboratory of Nutritional Epidemiology, School of Applied Sciences, University of Campinas (UNICAMP), Limeira 13484-350, São Paulo, Brazil
| | - Maria de Lourdes Setsuko Ayrizono
- IBD Research Laboratory, Colorectal Surgery Unit, School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-878, São Paulo, Brazil
| | - Marciane Milanski
- IBD Research Laboratory, Colorectal Surgery Unit, School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-878, São Paulo, Brazil
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas (UNICAMP), Campinas 13083-878, São Paulo, Brazil
| | - Raquel Franco Leal
- IBD Research Laboratory, Colorectal Surgery Unit, School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-878, São Paulo, Brazil
| |
Collapse
|
27
|
Long-Chain Polyunsaturated Fatty Acids (LCPUFAs) and the Developing Immune System: A Narrative Review. Nutrients 2021; 13:nu13010247. [PMID: 33467123 PMCID: PMC7830895 DOI: 10.3390/nu13010247] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/10/2021] [Accepted: 01/13/2021] [Indexed: 12/12/2022] Open
Abstract
The immune system is complex: it involves many cell types and numerous chemical mediators. An immature immune response increases susceptibility to infection, whilst imbalances amongst immune components leading to loss of tolerance can result in immune-mediated diseases including food allergies. Babies are born with an immature immune response. The immune system develops in early life and breast feeding promotes immune maturation and protects against infections and may protect against allergies. The long-chain polyunsaturated fatty acids (LCPUFAs) arachidonic acid (AA) and docosahexaenoic acid (DHA) are considered to be important components of breast milk. AA, eicosapentaenoic acid (EPA) and DHA are also present in the membranes of cells of the immune system and act through multiple interacting mechanisms to influence immune function. The effects of AA and of mediators derived from AA are often different from the effects of the n-3 LCPUFAs (i.e., EPA and DHA) and of mediators derived from them. Studies of supplemental n-3 LCPUFAs in pregnant women show some effects on cord blood immune cells and their responses. These studies also demonstrate reduced sensitisation of infants to egg, reduced risk and severity of atopic dermatitis in the first year of life, and reduced persistent wheeze and asthma at ages 3 to 5 years, especially in children of mothers with low habitual intake of n-3 LCPUFAs. Immune markers in preterm and term infants fed formula with AA and DHA were similar to those in infants fed human milk, whereas those in infants fed formula without LCPUFAs were not. Infants who received formula plus LCPUFAs (both AA and DHA) showed a reduced risk of allergic disease and respiratory illness than infants who received standard formula. Studies in which infants received n-3 LCPUFAs report immune differences from controls that suggest better immune maturation and they show lower risk of allergic disease and respiratory illness over the first years of life. Taken together, these findings suggest that LCPUFAs play a role in immune development that is of clinical significance, particularly with regard to allergic sensitisation and allergic manifestations including wheeze and asthma.
Collapse
|
28
|
Zheng L, Wen XL. Gut microbiota and inflammatory bowel disease: The current status and perspectives. World J Clin Cases 2021; 9:321-333. [PMID: 33521100 PMCID: PMC7812881 DOI: 10.12998/wjcc.v9.i2.321] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/20/2020] [Accepted: 12/06/2020] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic immune-mediated disease that affects the gastrointestinal tract. It is argued that environment, microbiome, and immune-mediated factors interact in a genetically susceptible host to trigger IBD. Recently, there has been increased interest in the development, progression, and treatment of IBD because of our understanding of the microbiome. Researchers have proved that some factors can alter the microbiome and the pathogenesis of IBD. As a result, there has been increasing interest in the application of probiotics, prebiotics, antibiotics, fecal microbiota transplantation, and gene manipulation in treating IBD because of the possible curative effect of microbiome-modulating interventions. In this review, we summarize the findings from human and animal studies and discuss the effect of the gut microbiome in treating patients with IBD.
Collapse
Affiliation(s)
- Lie Zheng
- Department of Gastroenterology, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi’an 730000, Shaanxi Province, China
| | - Xin-Li Wen
- Department of Gastroenterology, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi’an 730000, Shaanxi Province, China
| |
Collapse
|
29
|
Knežević D, Petković M. Faecal transplantation and Clostridioides difficile infection. SCRIPTA MEDICA 2021. [DOI: 10.5937/scriptamed52-32752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Faecal microbiota transplantation (FMT), known equally well as faecal transplantation or faecal bacteriotherapy, is the process of implanting the faecal suspension containing balanced microbiota from a healthy donor to the colon of a recipient patient. Excessive growth of Clostridioides difficile (C difficile) in the intestinal microbiota resulting from antibiotic consumption is currently a rising threat to public health. FMT is one of the most important, newer approaches to treating C difficile infections. Since C difficile is regarded as an opportunistic bacterium triggering disease in conditions of disturbed homeostasis of the intestinal microbiota, restoration of healthy intestinal microflora facilitates suppression of toxic strain of C difficile by anaerobic bacteria of normal intestinal microflora with concomitant cure. Nurses have important role in caring for patients after faecal transplantation.
Collapse
|
30
|
Abstract
Ulcerative colitis (UC) is a complex chronic, immune-mediated inflammatory disorder of the colon. Factors associated with increased risk of UC include diet, particularly Western diet influences in newly industrialized nations, medications, and lifestyle factors that may influence the host's microbiome or immune response to antigens. Although much evidence identifying potential genetic and host-related factors is currently available, there are still many unanswered questions. As the global UC incidence and prevalence continues to increase, there are multiple opportunities for continued investigation to clarify our understanding of UC, identify potential predictors of disease severity, response to therapy, and novel therapeutic targets.
Collapse
Affiliation(s)
- Lillian Du
- Inflammatory Bowel Diseases Center, Cedars-Sinai, 8730 Alden Drive Suite E204, Los Angeles, CA, USA
| | - Christina Ha
- Inflammatory Bowel Diseases Center, Cedars-Sinai, 8730 Alden Drive Suite E204, Los Angeles, CA, USA.
| |
Collapse
|
31
|
Czosnykowska-Łukacka M, Lis-Kuberka J, Królak-Olejnik B, Orczyk-Pawiłowicz M. Changes in Human Milk Immunoglobulin Profile During Prolonged Lactation. Front Pediatr 2020; 8:428. [PMID: 32850542 PMCID: PMC7426452 DOI: 10.3389/fped.2020.00428] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 06/19/2020] [Indexed: 01/09/2023] Open
Abstract
Mother's milk immunoglobulins (Igs) delivered to infants during breastfeeding are crucial in shaping and modulating immature infants' immune system and provide efficient protection against pathogens. The aim of the study was to evaluate the immunoglobulin concentrations in milk of 116 lactating mothers over prolonged lactation from the 1st to the 48th month using the ELISA method. The concentration of proteins, SIgA and IgG, but not IgM, showed a positive correlation (r = 0.69, p < 0.005; r = 0.54, p < 0.05; and r = 0.27, p < 0.05, respectively) with lactation from the 1st to the 48th month. The lowest concentrations of SIgA and IgG were observed for the first year (2.12 ± 0.62 g/L and 14.71 ± 6.18 mg/L, respectively) and the highest after the 2nd year of lactation (7.55 ± 7.16 g/L and 18.95 ± 6.76 mg/L, respectively). The IgM concentration remained stable during 2 years (2.81 ± 2.74 mg/L), but after 24 months it was higher (3.82 ± 3.05 mg/L), although not significantly. Moreover, negative correlations of protein (r = -0.24, p < 0.05) and SIgA (r = -0.47, p < 0.05) concentrations with the number of feedings were found. Human milk after the 2nd year of lactation contains significantly higher concentrations of protein, SIgA, and IgG. High concentration of immunoglobulins and protein during prolonged lactation is an additional argument to support breastfeeding even after introducing solid foods and should be one of the overarching goals in the protection of children's health.
Collapse
Affiliation(s)
| | - Jolanta Lis-Kuberka
- Department of Chemistry and Immunochemistry, Wroclaw Medical University, Wrocław, Poland
| | | | | |
Collapse
|
32
|
Melandri M, Aiudi GG, Caira M, Alonge S. A Biotic Support During Pregnancy to Strengthen the Gastrointestinal Performance in Puppies. Front Vet Sci 2020; 7:417. [PMID: 32851009 PMCID: PMC7417339 DOI: 10.3389/fvets.2020.00417] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/11/2020] [Indexed: 12/24/2022] Open
Abstract
Up to 60% of neonates can be affected by gastroenteritis due to specific pathogens or aspecific polymicrobial interactions. The present study evaluated if a dietary supplementation with MOS, FOS, E. faecium and L. acidophilus in pregnancy may reduce gastroenteritis in puppies. Fifteen Great Danes were divided in 3 groups. The control group (CG) ate a standard diet. In 2 study groups, the diet was supplemented with pre- and probiotics during the last (1WG) and the last 4 pregnancy weeks (4WG). Up to 9 weeks, puppies were checked daily to identify first- or second- presentation gastroenteritis. Data were processed by χ2 (P < 0.05). First-presentation gastroenteritis was more frequent in CG than in 1WG than in 4WG. Second-presentation gastroenteritis was more frequent in CG than in 1 and 4WG. Puppies from pre- and probiotics supplemented bitches were less prone to gastroenteritis. 1 or 4WG equally reduced second-presentation gastroenteritis in puppies, but 4WG was better than 1WG on first-presentation gastroenteritis. By entero-mammary link, supplemented bitches produced higher immune quality colostrum, thus puppies faced immunitary challenges better; moreover, maternal microbiota, positively altered by supplementation, was transferred to newborns, becoming more resistant to gastroenteritis. This information can be useful in clinical practice with the goal of preventing gastroenteritis in puppies and reducing its prevalence and severity.
Collapse
Affiliation(s)
| | - Giulio Guido Aiudi
- Dipartimento di Medicina Veterinaria, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Michele Caira
- Dipartimento di Medicina Veterinaria, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | | |
Collapse
|
33
|
Glassner KL, Abraham BP, Quigley EMM. The microbiome and inflammatory bowel disease. J Allergy Clin Immunol 2020; 145:16-27. [PMID: 31910984 DOI: 10.1016/j.jaci.2019.11.003] [Citation(s) in RCA: 535] [Impact Index Per Article: 107.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic immune-mediated disease affecting the gastrointestinal tract. IBD consists of 2 subtypes: ulcerative colitis and Crohn disease. IBD is thought to develop as a result of interactions between environmental, microbial, and immune-mediated factors in a genetically susceptible host. Of late, the potential role of the microbiome in the development, progression, and treatment of IBD has been a subject of considerable interest and enquiry. Indeed, studies in human subjects have shown that the gut microbiome is different in patients with IBD compared with that in healthy control subjects. Other evidence in support of a fundamental role for the microbiome in patients with IBD includes identification of mutations in genes involved in microbiome-immune interactions among patients with IBD and epidemiologic observations implicating such microbiota-modulating risk factors as antibiotic use, cigarette smoking, levels of sanitation, and diet in the pathogenesis of IBD. Consequently, there has been much interest in the possible benefits of microbiome-modulating interventions, such as probiotics, prebiotics, antibiotics, fecal microbiota transplantation, and gene manipulation in the treatment of IBD. In this review we will discuss the role of the gut microbiome in patients with IBD; our focus will be on human studies.
Collapse
Affiliation(s)
- Kerri L Glassner
- Fondren IBD Program, Lynda K. and David M. Underwood Center for Digestive Disorders, Division of Gastroenterology and Hepatology, Houston Methodist Hospital and Weill Cornell Medical College, Houston, Tex.
| | - Bincy P Abraham
- Fondren IBD Program, Lynda K. and David M. Underwood Center for Digestive Disorders, Division of Gastroenterology and Hepatology, Houston Methodist Hospital and Weill Cornell Medical College, Houston, Tex
| | - Eamonn M M Quigley
- Fondren IBD Program, Lynda K. and David M. Underwood Center for Digestive Disorders, Division of Gastroenterology and Hepatology, Houston Methodist Hospital and Weill Cornell Medical College, Houston, Tex
| |
Collapse
|
34
|
Saso A, Kampmann B. Maternal Immunization: Nature Meets Nurture. Front Microbiol 2020; 11:1499. [PMID: 32849319 PMCID: PMC7396522 DOI: 10.3389/fmicb.2020.01499] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/09/2020] [Indexed: 12/17/2022] Open
Abstract
Vaccinating women in pregnancy (i.e., maternal immunization) has emerged as a promising tool to tackle infant morbidity and mortality worldwide. This approach nurtures a 'gift of nature,' whereby antibody is transferred from mother to fetus transplacentally during pregnancy, or postnatally in breast milk, thereby providing passive, antigen-specific protection against infections in the first few months of life, a period of increased immune vulnerability for the infant. In this review, we briefly summarize the rationale for maternal immunization programs and the landscape of vaccines currently in use or in the pipeline. We then direct the focus to the underlying biological phenomena, including the main mechanisms by which maternally derived antibody is transferred efficiently to the infant, at the placental interface or in breast milk; important research models and methodological approaches to interrogate these processes, particularly in the context of recent advances in systems vaccinology; the potential biological and clinical impact of high maternal antibody titres on neonatal ontogeny and subsequent infant vaccine responses; and key vaccine- and host-related factors influencing the maternal-infant dyad across different environments. Finally, we outline important gaps in knowledge and suggest future avenues of research on this topic, proposing potential strategies to ensure optimal testing, delivery and implementation of maternal vaccination programs worldwide.
Collapse
Affiliation(s)
- Anja Saso
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Vaccines and Immunity Theme, MRC Unit The Gambia at LSHTM, Banjul, Gambia
| | - Beate Kampmann
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Vaccines and Immunity Theme, MRC Unit The Gambia at LSHTM, Banjul, Gambia
| |
Collapse
|
35
|
Microbial biodiversity in the throats of pulmonary tuberculosis patients and tuberculin skin test (TST) positive and negative healthy individuals in Malaysia. Tuberculosis (Edinb) 2020; 124:101965. [PMID: 32692651 DOI: 10.1016/j.tube.2020.101965] [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: 02/19/2020] [Revised: 06/18/2020] [Accepted: 06/26/2020] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to investigate the composition of throat microbiota in pulmonary tuberculosis patients (PTB) in comparison to healthy tuberculin skin test positive (TSTp) and negative (TSTn) individuals. Throat swabs samples were collected, and the microbiota was characterized. Richer operational taxonomic units (OTUs) were present in PTB group, compared to TSTp and TSTn. Regarding alpha diversity analysis there was a higher community diversity in TSTn compared to TSTp. Beta diversity analysis showed different species composition in TSTp compared to TSTn and PTB. There was higher presence of Firmicutes in PTB and TSTn compared to TSTp group at phylum level. At the genus level, Leuconostoc and Enterococcus were higher in TSTn compared to TSTp and Pediococcus, Chryseobacterium, Bifidobacterium, Butyrivibrio, and Bulleidia were higher in PTB compared to TSTn. Streptococcus was higher in TSTn compared to PTB and Lactobacillus in PTB compared to TSTp. At species level, Streptococcus sobrinus and Bulleidia moorei were higher in PTB compared to TSTn individuals, while Lactobacillus salivarius was higher in PTB compared to TSTp. The differences in the microbiome composition could influence the resistance/susceptibility to Mtb infection.
Collapse
|
36
|
Kumar T, Pandey R, Chauhan NS. Hypoxia Inducible Factor-1α: The Curator of Gut Homeostasis. Front Cell Infect Microbiol 2020; 10:227. [PMID: 32500042 PMCID: PMC7242652 DOI: 10.3389/fcimb.2020.00227] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/23/2020] [Indexed: 12/11/2022] Open
Abstract
The human gut microbiome is a stratified and resilient ecosystem co-inhabited by a diverse and dynamic pool of microorganisms. Microbial selection, establishment, and colonization are modulated through a complex molecular network of host-microbial interactions. These molecular bioprocesses ensure the taxonomic composition of the mature human gut microbiome. The human gut microbiome plays a vital role in host health; otherwise, any microbial dysbiosis could predispose to the onset of physiological and metabolic disorder/s. Focussed research are being carried out to identify key molecular agents defining gut homeostasis. These molecules hold the potential to develop effective therapeutic solutions for microbial dysbiosis-associated human disorders. Of these, Hypoxia-inducible factor-1α (HIF-1α) is a central player in host-microbial crosstalk to maintain gut homeostasis. Human gut microbial metabolites regulate its cellular stability, which in turn regulates various cellular processes required for the stable gut microbiome. In the present review, an effort has been made to summarize the key role of HIF-1α to maintain gut homeostasis.
Collapse
Affiliation(s)
- Tarun Kumar
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, India
| | - Rajesh Pandey
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India
| | - Nar Singh Chauhan
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, India
| |
Collapse
|
37
|
Salehi B, Dimitrijević M, Aleksić A, Neffe-Skocińska K, Zielińska D, Kołożyn-Krajewska D, Sharifi-Rad J, Stojanović-Radić Z, Prabu SM, Rodrigues CF, Martins N. Human microbiome and homeostasis: insights into the key role of prebiotics, probiotics, and symbiotics. Crit Rev Food Sci Nutr 2020; 61:1415-1428. [PMID: 32400169 DOI: 10.1080/10408398.2020.1760202] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The interest in the study of the gut microbiome has grown exponentially. Indeed, its impact on health and disease has been increasingly reported, and the importance of keeping gut microbiome homeostasis clearly highlighted. However, and despite many advances, there are still some gaps, as well as the real discernment on the contribution of some species falls far short of what is needed. Anyway, it is already more than a solid fact of its importance in maintaining health and preventing disease, as well as in the treatment of some pathologies. In this sense, and given the existence of some ambiguous opinions, the present review aims to discuss the importance of gut microbiome in homeostasis maintenance, and even the role of probiotics, prebiotics, and symbiotics in both health promotion and disease prevention.
Collapse
Affiliation(s)
- Bahare Salehi
- Student Research Committee, School of Medicine, Bam University of Medical Sciences, Bam, Iran
| | - Marina Dimitrijević
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | - Ana Aleksić
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | - Katarzyna Neffe-Skocińska
- Department of Food Gastronomy and Food Hygiene, Warsaw University of Life Sciences (WULS), Warszawa, Poland
| | - Dorota Zielińska
- Department of Food Gastronomy and Food Hygiene, Warsaw University of Life Sciences (WULS), Warszawa, Poland
| | - Danuta Kołożyn-Krajewska
- Department of Food Gastronomy and Food Hygiene, Warsaw University of Life Sciences (WULS), Warszawa, Poland
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zorica Stojanović-Radić
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | | | - Célia F Rodrigues
- LEPABE - Department of Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Natália Martins
- Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, Porto, Portugal.,Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal
| |
Collapse
|
38
|
Abstract
Human milk provides not only ideal nutrition for infant development but also immunologic factors to protect from infection and inflammation. For the newborn preterm infant, the natural delivery of milk is not attainable, and instead pumped maternal milk, donor human milk, and human milk fortification are mainstays of clinical care. Current research demonstrates a decreased risk of necrotizing enterocolitis with maternal milk and donor human milk when individually compared to formula and with a complete human milk diet of maternal milk supplemented with donor human milk. The incidence of severe retinopathy of prematurity is decreased with an exclusive human milk diet, and this decrease is more pronounced with human milk-based compared to bovine milk-based human milk fortifier. The incidence of other morbidities such as late-onset sepsis and bronchopulmonary dysplasia is decreased with higher dose of human milk though significant differences are not apparent in exclusive human milk diet studies.
Collapse
Affiliation(s)
- Sarah N Taylor
- Yale School of Medicine, PO Box 208064, New Haven, Connecticut, 06520-8064, USA.
| |
Collapse
|
39
|
Chastant S, Mila H. Passive immune transfer in puppies. Anim Reprod Sci 2019; 207:162-170. [PMID: 31255495 PMCID: PMC7125514 DOI: 10.1016/j.anireprosci.2019.06.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/12/2019] [Accepted: 06/12/2019] [Indexed: 12/20/2022]
Abstract
The puppy, born without immunoglobulins G (IgG), acquires a passive systemic immunity thanks to colostrum intake during the two first days of life. The quality of passive immune transfer (i.e. blood IgG concentration at two days of age), highly variable between litters and between puppies within litters, depends mainly on the time elapsed between birth and ingestion of colostrum, with limited influence of colostrum IgG concentration. Deficit in passive immune transfer, impacting puppy's health and neonatal mortality rate, can be indirectly diagnosed through blood gammaglutamyltransferases assay and evaluation of growth rate over the two first days of life. In the absence of maternal colostrum, few homo- and heterospecific immune sources are available and canine colostrum banking remains the optimal solution. Whereas passive immune transfer is crucial for survival during the neonatal period, it later interferes with response to vaccination. In addition to systemic passive immune transfer, maternal antibodies (mainly IgA) would provide local (digestive) immunity, ensuring mid-term protection of the puppies' gut together with probably long term training of the digestive immune system.
Collapse
Affiliation(s)
- Sylvie Chastant
- NeoCare, UMR INRA/ENVT 1225 Interactions Host-Pathogens, Toulouse National Veterinary School, 23 Chemin des CAPELLES, BP 87614, 31076, Toulouse Cedex, France.
| | - Hanna Mila
- NeoCare, UMR INRA/ENVT 1225 Interactions Host-Pathogens, Toulouse National Veterinary School, 23 Chemin des CAPELLES, BP 87614, 31076, Toulouse Cedex, France.
| |
Collapse
|
40
|
Wang Q, Fu W, Guo Y, Tang Y, Du H, Wang M, Liu Z, Li Q, An L, Tian J, Li M, Wu Z. Drinking Warm Water Improves Growth Performance and Optimizes the Gut Microbiota in Early Postweaning Rabbits during Winter. Animals (Basel) 2019; 9:E346. [PMID: 31212853 PMCID: PMC6616395 DOI: 10.3390/ani9060346] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/04/2019] [Accepted: 06/07/2019] [Indexed: 12/31/2022] Open
Abstract
Accumulating evidence indicates that cold exposure changes the composition of the gut microbiota and reduces intestinal immunity in early postweaning livestock. However, little is known about the effects of drinking warm water (WW) on gut microbiota during winter. In this study, we investigated the effects of drinking WW in winter on the growth performance and gut microbiota structure of rabbits raised in poorly insulated housing from the early postweaning period (day 46) to the subadult period (day 82). The average daily gain and feed conversion ratio in rabbits drinking WW were significantly improved compared to those of the rabbits drinking cold water (CW) during 47-58 days. In addition, rabbits drinking WW had a significantly decreased the risk of diarrhea during 71-82 days. 16S rRNA sequence analysis revealed that the alpha diversity of the cecal microbiota was not significantly different between the WW and CW groups, but significantly increased with age. The relative abundance of cecal microorganisms, such as Coprococcus spp. was considerably increased at day 70 in the group drinking WW. Correlation analysis indicated that Coprococcus spp. was negatively associated with pro-inflammatory factors. In conclusion, our results suggest that drinking WW has a positive effect on growth performance and gut microbiota in rabbits during the early postweaning stage in winter.
Collapse
Affiliation(s)
- Qiangjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Wei Fu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Yao Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Yuhan Tang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
- ZhaoTong Technology Promotion Workstation of Animal Husbandry and Veterinary Medicine, ZhaoTong 657000, China.
| | - Haoxuan Du
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Meizhi Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Zhongying Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Qin Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Lei An
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Jianhui Tian
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Mingyong Li
- National Rabbit Industry Technology System Qingdao Comprehensive Experimental Station, Qingdao 266431, China.
| | - Zhonghong Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
41
|
Levi Mortera S, Soggiu A, Vernocchi P, Del Chierico F, Piras C, Carsetti R, Marzano V, Britti D, Urbani A, Roncada P, Putignani L. Metaproteomic investigation to assess gut microbiota shaping in newborn mice: A combined taxonomic, functional and quantitative approach. J Proteomics 2019; 203:103378. [PMID: 31102759 DOI: 10.1016/j.jprot.2019.103378] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/23/2019] [Accepted: 05/13/2019] [Indexed: 12/16/2022]
Abstract
Breastfeeding is nowadays known to be one of the most critical factors contributing to the development of an efficient immune system. In the last decade, a consistent number of pieces of evidence demonstrated the relationship between a healthy organism and its gut microbiota. However, this link is still not fully understood and requires further investigation. We recently adopted a murine model to describe the impact of either maternal milk or parental genetic background, on the composition of the gut microbial population in the first weeks of life. A metaproteomic approach to such complex environments is a big challenge that requires a strong effort in both data production and analysis, including the set-up of dedicated multitasking bioinformatics pipelines. Herein we present an LC-MS/MS based investigation to monitor mouse gut microbiota in the early life, aiming at characterizing its functions and metabolic activities together with a taxonomic description in terms of operational taxonomic units. We provided a quantitative evaluation of bacterial metaproteins, taking into account differential expression results in relation to the functional and taxonomic classification, particularly with proteins from orthologues groups. This allowed the reduction of the bias arising from the presence of a high number of shared peptides, and proteins, among different bacterial species. We also focused on host mucosal proteome and its modulation, according to different microbiota composition. SIGNIFICANCE: This paper would represent a reference work for investigations on gut microbiota in early life, from both a microbiological and a functional proteomic point of view. We focused on the shaping of the mouse gut microbiota in dependence on the feeding modality, defining a reliable taxonomic description, highlighting some functional characteristics of the microbial community, and performing a first quantitative evaluation by data independent analysis in metaproteomics.
Collapse
Affiliation(s)
| | - Alessio Soggiu
- Department of Veterinary Medicine, University of Milan, Milan, Italy
| | - Pamela Vernocchi
- Human Microbiome Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Cristian Piras
- Department of Veterinary Medicine, University of Milan, Milan, Italy
| | - Rita Carsetti
- B cell Pathophysiology Unit, Immunology Research Area and Unit of Diagnostic Immunology, Department of Laboratories, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Valeria Marzano
- Human Microbiome Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Domenico Britti
- C.I.S. - Interdepartmental Services Centre of Veterinary for Human and Animal Health, University of Catanzaro "Magna Græcia", Catanzaro, Italy.; Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Andrea Urbani
- Catholic University of Sacred Heart, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Paola Roncada
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Lorenza Putignani
- Parasitology Unit and Human Microbiome Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| |
Collapse
|
42
|
Adipositas, metabolische Komorbiditäten und Mikrobiommodulation in der Pädiatrie. Monatsschr Kinderheilkd 2019. [DOI: 10.1007/s00112-019-0689-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
43
|
McKeen S, Young W, Mullaney J, Fraser K, McNabb WC, Roy NC. Infant Complementary Feeding of Prebiotics for theMicrobiome and Immunity. Nutrients 2019; 11:nu11020364. [PMID: 30744134 PMCID: PMC6412789 DOI: 10.3390/nu11020364] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/01/2019] [Accepted: 02/06/2019] [Indexed: 02/06/2023] Open
Abstract
Complementary feeding transitions infants from a milk-based diet to solid foods, providing essential nutrients to the infant and the developing gut microbiome while influencing immune development. Some of the earliest microbial colonisers readily ferment select oligosaccharides, influencing the ongoing establishment of the microbiome. Non-digestible oligosaccharides in prebiotic-supplemented formula and human milk oligosaccharides promote commensal immune-modulating bacteria such as Bifidobacterium, which decrease in abundance during weaning. Incorporating complex, bifidogenic, non-digestible carbohydrates during the transition to solid foods may present an opportunity to feed commensal bacteria and promote balanced concentrations of beneficial short chain fatty acid concentrations and vitamins that support gut barrier maturation and immunity throughout the complementary feeding window.
Collapse
Affiliation(s)
- Starin McKeen
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Wayne Young
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Jane Mullaney
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Karl Fraser
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Warren C McNabb
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Nicole C Roy
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| |
Collapse
|
44
|
Klein LD, Huang J, Quinn EA, Martin MA, Breakey AA, Gurven M, Kaplan H, Valeggia C, Jasienska G, Scelza B, Lebrilla CB, Hinde K. Variation among populations in the immune protein composition of mother's milk reflects subsistence pattern. Evol Med Public Health 2018; 2018:230-245. [PMID: 30430010 PMCID: PMC6222208 DOI: 10.1093/emph/eoy031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 09/25/2018] [Indexed: 12/29/2022] Open
Abstract
LAY SUMMARY Adaptive immune proteins in mothers' milk are more variable than innate immune proteins across populations and subsistence strategies. These results suggest that the immune defenses in milk are shaped by a mother's environment throughout her life. BACKGROUND AND OBJECTIVES Mother's milk contains immune proteins that play critical roles in protecting the infant from infection and priming the infant's developing immune system during early life. The composition of these molecules in milk, particularly the acquired immune proteins, is thought to reflect a mother's immunological exposures throughout her life. In this study, we examine the composition of innate and acquired immune proteins in milk across seven populations with diverse disease and cultural ecologies. METHODOLOGY Milk samples (n = 164) were collected in Argentina, Bolivia, Nepal, Namibia, Philippines, Poland and the USA. Populations were classified as having one of four subsistence patterns: urban-industrialism, rural-shop, horticulturalist-forager or agro-pastoralism. Milk innate (lactalbumin, lactoferrin and lysozyme) and acquired (Secretory IgA, IgG and IgM) protein concentrations were determined using triple-quadrupole mass spectrometry. RESULTS Both innate and acquired immune protein composition in milk varied among populations, though the acquired immune protein composition of milk differed more among populations. Populations living in closer geographic proximity or having similar subsistence strategies (e.g. agro-pastoralists from Nepal and Namibia) had more similar milk immune protein compositions. Agro-pastoralists had different milk innate immune protein composition from horticulturalist-foragers and urban-industrialists. Acquired immune protein composition differed among all subsistence strategies except horticulturist-foragers and rural-shop. CONCLUSIONS AND IMPLICATIONS Our results reveal fundamental variation in milk composition that has not been previously explored in human milk research. Further study is needed to understand what specific aspects of the local environment influence milk composition and the effects this variation may have on infant health outcomes.
Collapse
Affiliation(s)
- Laura D Klein
- Department of Human Evolutionary Biology, Harvard University, 11 Divinity Avenue, Cambridge, MA, USA
- Department of Anthropology, University of Illinois at Chicago, 1007 West Harrison Street, Chicago IL, USA
| | - Jincui Huang
- Chemistry Department, University of California Davis, 2465 Chemistry Annex, One Shields Avenue, Davis, CA, USA
| | - Elizabeth A Quinn
- Department of Anthropology, Washington University in St Louis, Campus Box 1114, One Brookings Drive, St Louis, MO, USA
| | - Melanie A Martin
- Department of Anthropology, University of California Santa Barbara, Santa Barbara, CA, USA
- Department of Anthropology, University of Washington, 314 Denny Hall, Box 353100, Seattle, WA, USA
| | - Alicia A Breakey
- Department of Human Evolutionary Biology, Harvard University, 11 Divinity Avenue, Cambridge, MA, USA
| | - Michael Gurven
- Department of Anthropology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Hillard Kaplan
- Department of Anthropology, University of New Mexico, MSC01-1040, 1 University of New Mexico, Albuquerque, NM, USA
| | - Claudia Valeggia
- Department of Anthropology, Yale University, 10 Sachem Street, New Haven, CT, USA
| | - Grazyna Jasienska
- Department of Environmental Health, Faculty of Health Sciences, Jagiellonian University Medical College, ul. Grzegorzecka 20, Krakow, Poland
| | - Brooke Scelza
- Department of Anthropology, University of California Los Angeles, 341 Haines Hall, Box 951553, Los Angeles, CA, USA
| | - Carlito B Lebrilla
- Chemistry Department, University of California Davis, 2465 Chemistry Annex, One Shields Avenue, Davis, CA, USA
| | - Katie Hinde
- Department of Human Evolutionary Biology, Harvard University, 11 Divinity Avenue, Cambridge, MA, USA
- School of Human Evolution and Social Change
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
45
|
Riaz Rajoka MS, Zhao H, Li N, Lu Y, Lian Z, Shao D, Jin M, Li Q, Zhao L, Shi J. Origination, change, and modulation of geriatric disease-related gut microbiota during life. Appl Microbiol Biotechnol 2018; 102:8275-8289. [PMID: 30066188 DOI: 10.1007/s00253-018-9264-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/21/2018] [Accepted: 07/26/2018] [Indexed: 12/21/2022]
Abstract
The age-related changes in the diversity and composition of the gut microbiota are well described in recent studies. These changes have been suggested to be influenced by age-associated weakening of the immune system and low-grade chronic inflammation, resulting in numerous age-associated pathological conditions. Gut microbiota homeostasis is important throughout the life of the host by providing vital functions to regulate various immunological functions and homeostasis. Based on published results, we summarize the relationship between the gut microbiota and aging-related diseases, especially Parkinson's disease, immunosenescence, rheumatoid arthritis, bone loss, and metabolic syndrome. The change in composition of the gut microbiota and gut ecosystem during life and its influence on the host immunologic and metabolic phenotype are also analyzed to determine factors that affect aging-related diseases. Approaches to maintain host health and prevent or cure geriatric diseases are also discussed.
Collapse
Affiliation(s)
- Muhammad Shahid Riaz Rajoka
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
- Department of Food Science and Engineering, College of Chemistry and Chemical Engineering, Shenzhen University, Shenzhen, Guangdong, People's Republic of China
| | - Haobin Zhao
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Na Li
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Yao Lu
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Ziyang Lian
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Dongyan Shao
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Mingliang Jin
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Qi Li
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Liqing Zhao
- Department of Food Science and Engineering, College of Chemistry and Chemical Engineering, Shenzhen University, Shenzhen, Guangdong, People's Republic of China
| | - Junling Shi
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China.
| |
Collapse
|
46
|
The Effect of Early-Life Environmental Exposures on Disease Phenotype and Clinical Course of Crohn's Disease in Children. Am J Gastroenterol 2018; 113:1524-1529. [PMID: 30267029 DOI: 10.1038/s41395-018-0239-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/29/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Environmental factors play an important role in the pathogenesis of Crohn's Disease (CD). In particular, by virtue of the instability of the microbiome and development of immunologic tolerance, early life factors may exert the strongest influence on disease risk and phenotype. METHODS We used data from 1119 CD subjects recruited from RISK inception cohort to examine the impact of early life environment on disease progression. Our primary exposures of interest were breastfeeding in infancy and exposure to maternal, active, or passive smoke. Our primary outcomes were development of complicated (stricturing or penetrating) disease, and need for CD-related hospitalization, and surgery. Multivariable logistic regression models were used to define independent associations, adjusting for relevant covariates. RESULTS Our study cohort included 1119 patients with CD among whom 15% had stricturing (B2) or penetrating disease (B3) by 3 years. 331 patients (35%) and 95 patients (10.6%) required CD-related hospitalizations and surgery respectively. 74.5% were breastfed in infancy and 31% were exposed to smoking among whom 7% were exposed to maternal smoke. On multivariable analysis, a history of breastfeeding was inversely associated with complicated (B2/B3 disease) 0.65, CI 95% 0.44-96; P = 0.03) in pediatric CD. Maternal smoking during pregnancy was associated with increased risk of hospitalization during the 3-year follow-up period (OR 1.75, CI 95% 1.05-2.89; P = 0.03). CONCLUSIONS Early life environmental factors influence the eventual phenotypes and disease course in CD.
Collapse
|
47
|
Sakaguchi K, Koyanagi A, Kamachi F, Harauma A, Chiba A, Hisata K, Moriguchi T, Shimizu T, Miyake S. Breast-feeding regulates immune system development via transforming growth factor-β in mice pups. Pediatr Int 2018; 60:224-231. [PMID: 29290091 DOI: 10.1111/ped.13507] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 06/17/2017] [Accepted: 10/31/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND Breast milk contains important nutrients and immunoregulatory factors that are essential for newborn infants. Recently, epidemiological studies suggested that breast-feeding prevents a wide range of infectious diseases and lowers the incidence of infant allergic diseases. METHODS To examine the effects of breast milk on immunological development in infancy, we established an artificial rearing system for hand-feeding mice and compared mouse pups fed with either breast milk or milk substitute. All mice were killed at 14 days of age and immune cells in the thymus, spleen, and small intestine were examined on flow cytometry. RESULTS The number of thymocytes was higher whereas that of total immune cells of peripheral lymphoid tissues was lower in mice fed breast milk compared with milk substitute-fed mice. In peripheral lymphoid tissues, the proportion of B cells was higher and that of CD8+ T cells, macrophages, dendritic cells, and granulocytes was significantly lower in breast milk-fed mice. The same alteration in immune cells of the thymus and peripheral lymphoid tissues in milk substitute-fed mice was also observed in pups reared by mother mice treated with anti-transforming growth factor-β (anti-TGF-β) monoclonal antibody. CONCLUSIONS Breast milk regulates the differentiation and expansion of innate and adaptive immune cells partly due to TGF-β. Hence, TGF-β in breast milk may be a new therapeutic target for innate immune system-mediated diseases of infancy.
Collapse
Affiliation(s)
- Keita Sakaguchi
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akemi Koyanagi
- Laboratory of Cell Biology, Juntendo University Graduate School of Medicine Research Center, Tokyo, Japan
| | - Fumitaka Kamachi
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Akiko Harauma
- School of Life and Environmental Science, Azabu University, Sagamihara, Kanagawa, Japan
| | - Asako Chiba
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Ken Hisata
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Toru Moriguchi
- School of Life and Environmental Science, Azabu University, Sagamihara, Kanagawa, Japan
| | - Toshiaki Shimizu
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Sachiko Miyake
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
48
|
Riaz Rajoka MS, Zhao H, Lu Y, Lian Z, Li N, Hussain N, Shao D, Jin M, Li Q, Shi J. Anticancer potential against cervix cancer (HeLa) cell line of probiotic Lactobacillus casei and Lactobacillus paracasei strains isolated from human breast milk. Food Funct 2018; 9:2705-2715. [DOI: 10.1039/c8fo00547h] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
The CFCS of Lactobacilli strains from human breast milk induced apoptosis in HeLa cells ROS related mitochondrial pathway.
Collapse
|
49
|
Xu L, Lochhead P, Ko Y, Claggett B, Leong RW, Ananthakrishnan AN. Systematic review with meta-analysis: breastfeeding and the risk of Crohn's disease and ulcerative colitis. Aliment Pharmacol Ther 2017; 46:780-789. [PMID: 28892171 PMCID: PMC5688338 DOI: 10.1111/apt.14291] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 07/24/2017] [Accepted: 08/16/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Breastfeeding is a modifiable factor that may influence development of inflammatory bowel diseases. However, literature on this has been inconsistent and not accounted for heterogeneity in populations and exposure. AIM To conduct a meta-analysis to examine the association between breastfeeding in infancy and risk of Crohn's disease (CD) and ulcerative colitis (UC). METHODS A systematic search of Medline/PubMed and Embase was performed for full text, English-language literature through November 2016. Studies were included if they described breastfeeding in infancy in patients with CD or UC, and healthy controls. Data were pooled using a random effects model for analysis. RESULTS A total of 35 studies were included in the final analysis, comprising 7536 individuals with CD, 7353 with UC and 330 222 controls. Ever being breastfed was associated with a lower risk of CD (OR 0.71, 95% CI 0.59-0.85) and UC (OR 0.78, 95% CI 0.67-0.91). While this inverse association was observed in all ethnicity groups, the magnitude of protection was significantly greater among Asians (OR 0.31, 95% CI 0.20-0.48) compared to Caucasians (OR 0.78, 95% CI 0.66-0.93; P = .0001) in CD. Breastfeeding duration showed a dose-dependent association, with strongest decrease in risk when breastfed for at least 12 months for CD (OR 0.20, 95% CI 0.08-0.50) and UC (OR 0.21, 95% CI 0.10-0.43) as compared to 3 or 6 months. CONCLUSION Breastfeeding in infancy protects against the development of CD and ulcerative colitis.
Collapse
Affiliation(s)
- Luyi Xu
- Division of Gastroenterology, Massachusetts General Hospital, Boston, United States,Harvard Medical School, Boston, United States
| | - Paul Lochhead
- Division of Gastroenterology, Massachusetts General Hospital, Boston, United States
| | - Yanna Ko
- Concord Hospital, Gastroenterology and Liver Services, Sydney, Australia
| | - Brian Claggett
- Harvard Medical School, Boston, United States,Cardiac Imaging Core Laboratory and Clinical Trials Endpoints Center, Brigham and Women’s Hospital, Boston, United States
| | - Rupert W Leong
- Concord Hospital, Gastroenterology and Liver Services, Sydney, Australia
| | - Ashwin N Ananthakrishnan
- Division of Gastroenterology, Massachusetts General Hospital, Boston, United States,Harvard Medical School, Boston, United States
| |
Collapse
|
50
|
Ramírez-Pérez O, Cruz-Ramón V, Chinchilla-López P, Méndez-Sánchez N. The Role of the Gut Microbiota in Bile Acid Metabolism. Ann Hepatol 2017; 16 Suppl 1:S21-S26. [PMID: 31196631 DOI: 10.5604/01.3001.0010.5672] [Citation(s) in RCA: 189] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 09/09/2017] [Indexed: 02/04/2023]
Abstract
The gut microbiota has been considered a cornerstone of maintaining the health status of its human host because it not only facilitates harvesting of nutrients and energy from ingested food, but also produces numerous metabolites that can regulate host metabolism. One such class of metabolites, the bile acids, are synthesized from cholesterol in the liver and further metabolized by the gut microbiota into secondary bile acids. These bioconversions modulate the signaling properties of bile acids through the nuclear farnesoid X receptor and the G protein-coupled membrane receptor 5, which regulate diverse metabolic pathways in the host. In addition, bile acids can regulate gut microbial composition both directly and indirectly by activation of innate immune response genes in the small intestine. Therefore, host metabolism can be affected by both microbial modifications of bile acids, which leads to altered signaling via bile acid receptors, and by alterations in the composition of the microbiota. In this review, we mainly describe the interactions between bile acids and intestinal microbiota and their roles in regulating host metabolism, but we also examine the impact of bile acid composition in the gut on the intestinal microbiome and on host physiology.
Collapse
Affiliation(s)
| | - Vania Cruz-Ramón
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| | | | | |
Collapse
|