1
|
Bianco I, Ferrara C, Romano F, Loperfido F, Sottotetti F, El Masri D, Vincenti A, Cena H, De Giuseppe R. The Influence of Maternal Lifestyle Factors on Human Breast Milk Microbial Composition: A Narrative Review. Biomedicines 2024; 12:2423. [PMID: 39594990 PMCID: PMC11592219 DOI: 10.3390/biomedicines12112423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/28/2024] Open
Abstract
Human breast milk (HBM) is considered the gold standard for infant nutrition due to its optimal nutrient profile and complex composition of cellular and non-cellular components. Breastfeeding positively influences the newborn's gut microbiota and health, reducing the risk of conditions like gastrointestinal infections and chronic diseases (e.g., allergies, asthma, diabetes, and obesity). Research has revealed that HBM contains beneficial microbes that aid gut microbiota maturation through mechanisms like antimicrobial production and pathogen exclusion. The HBM microbiota composition can be affected by several factors, including gestational age, delivery mode, medical treatments, lactation stage, as well as maternal lifestyle habits (e.g., diet, physical activity, sleep quality, smoking, alcohol consumption, stress level). Particularly, lifestyle factors can play a significant role in shaping the HBM microbiota by directly modulating the microbial composition or influencing the maternal gut microbiota and influencing the HBM microbes through the enteromammary pathway. This narrative review of current findings summarized how maternal lifestyle influences HBM microbiota. While the influence of maternal diet on HBM microbiota is well-documented, indicating that dietary patterns, especially those rich in plant-based proteins and complex carbohydrates, can positively influence HBM microbiota, the impact of other lifestyle factors is poorly investigated. Maintaining a healthy lifestyle during pregnancy and breastfeeding is crucial for the health of both mother and baby. Understanding how maternal lifestyle factors influence microbial colonization of HBM, along with their interactions and impact, is key to developing new strategies that support the beneficial maturation of the infant's gut microbiota.
Collapse
Affiliation(s)
- Irene Bianco
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
| | - Chiara Ferrara
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
| | - Francesca Romano
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
| | - Federica Loperfido
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
| | - Francesca Sottotetti
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
| | - Dana El Masri
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
| | - Alessandra Vincenti
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
| | - Hellas Cena
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
- Clinical Nutrition Unit, General Medicine, Istituti Clinici Scientifici (ICS) Maugeri, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 27100 Pavia, Italy
| | - Rachele De Giuseppe
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
| |
Collapse
|
2
|
Heinecke F, Fornes D, Capobianco E, Flores Quiroga JP, Labiano M, Faletti AG, Jawerbaum A, White V. Intestinal alterations and mild glucose homeostasis impairments in the offspring born to overweight rats. Mol Cell Endocrinol 2024; 587:112201. [PMID: 38494045 DOI: 10.1016/j.mce.2024.112201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024]
Abstract
The gut plays a crucial role in metabolism by regulating the passage of nutrients, water and microbial-derived substances to the portal circulation. Additionally, it produces incretins, such as glucose-insulinotropic releasing peptide (GIP) and glucagon-like derived peptide 1 (GLP1, encoded by gcg gene) in response to nutrient uptake. We aimed to investigate whether offspring from overweight rats develop anomalies in the barrier function and incretin transcription. We observed pro-inflammatory related changes along with a reduction in Claudin-3 levels resulting in increased gut-permeability in fetuses and offspring from overweight rats. Importantly, we found decreased gip mRNA levels in both fetuses and offspring from overweight rats. Differently, gcg mRNA levels were upregulated in fetuses, downregulated in female offspring and unchanged in male offspring from overweight rats. When cultured with high glucose, intestinal explants showed an increase in gip and gcg mRNA levels in control offspring. In contrast, offspring from overweight rats did not exhibit any response in gip mRNA levels. Additionally, while females showed no response, male offspring from overweight rats did exhibit an upregulation in gcg mRNA levels. Furthermore, female and male offspring from overweight rats showed sex-dependent anomalies when orally challenged with a glucose overload, returning to baseline glucose levels after 120 min. These results open new research questions about the role of the adverse maternal metabolic condition in the programming of impairments in glucose homeostasis, enteroendocrine function and gut barrier function in the offspring from overweight mothers and highlight the importance of a perinatal maternal healthy metabolism.
Collapse
Affiliation(s)
- Florencia Heinecke
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Daiana Fornes
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Evangelina Capobianco
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Jeremias Pablo Flores Quiroga
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Marina Labiano
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Alicia G Faletti
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Alicia Jawerbaum
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Verónica White
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
3
|
Bai T, Wang Z, Shao H, Zhang X, Lorenz A, Meng X, Wu Y, Chen H, Li X. Novel Perspective on the Regulation of Offspring Food Allergy by Maternal Diet and Nutrients. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:10679-10691. [PMID: 38695770 DOI: 10.1021/acs.jafc.3c09108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
There has been a dramatic surge in the prevalence of food allergy (FA) that cannot be explained solely by genetics, identifying mechanisms of sensitization that are driven by environmental factors has become increasingly important. Diet, gut microbiota, and their metabolites have been shown to play an important role in the development of FA. In this review, we discuss the latest epidemiological evidence on the impact of two major dietary patterns and key nutrients in early life on the risk of offspring developing FA. The Western diet typically includes high sugar and high fat, which may affect the immune system of offspring and increase susceptibility to FA. In contrast, the Mediterranean diet is rich in fiber, which may reduce the risk of FA in offspring. Furthermore, we explore the potential mechanisms by which maternal dietary nutrients during a window of opportunity (pregnancy, birth, and lactation) influences the susceptibility of offspring to FA through multi-interface crosstalk. Finally, we discuss the limitations and gaps in the available evidence regarding the relationship between maternal dietary nutrients and the risk of FA in offspring. This review provides novel perspective on the regulation of offspring FA by maternal diet and nutrients.
Collapse
Affiliation(s)
- Tianliang Bai
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
- School of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Zhongliang Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
- School of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Huming Shao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
- School of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Xing Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
- School of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Agla Lorenz
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg 5020, Austria
| | - Xuanyi Meng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi 330047, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Yong Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi 330047, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi 330047, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Xin Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
- School of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi 330047, China
| |
Collapse
|
4
|
Kebbe M, Shankar K, Redman LM, Andres A. Human Milk Components and the Infant Gut Microbiome at 6 Months: Understanding the Interconnected Relationship. J Nutr 2024; 154:1200-1208. [PMID: 38442855 DOI: 10.1016/j.tjnut.2024.02.029] [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: 12/18/2023] [Revised: 01/29/2024] [Accepted: 02/26/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Human milk oligosaccharides have been shown to relate to the infant gut microbiome. However, the impact of other human milk components on infant gut bacterial colonization remains unexplored. OBJECTIVES Our cross-sectional analysis aimed to investigate associations between human milk components (energy, macronutrients, free amino acids, inflammatory markers, and hormones) and infant gut microbiome diversity and composition (phylum, family, and genus) at 6 mo of age. METHODS Human milk and infant stool samples were collected at 6 mo postpartum. The infant gut microbiome was profiled using 16S rRNA sequencing. Linear regression models were performed to examine associations, adjusting for pregravid BMI (kg/m2), delivery mode, duration of human milk feeding, and infant sex, with q < 0.2 considered significant. RESULTS This analysis included a total of 54 mothers (100% exclusively feeding human milk) and infants (n = 28 male; 51.9%). Total energy in human milk showed a negative association with α-diversity measures (Chao1 and Shannon). Interleukin (IL)-8 in human milk was positively associated with Chao1 and observed operational taxonomic units. At the family level, human milk glutamine and serine levels showed a negative association with the abundance of Veillonellaceae, whereas isoleucine showed a positive association with Bacteroidaceae. Human milk IL-8 and IL-6 concentrations were positively associated with Bacteroidaceae abundance. IL-8 also had a positive relationship with Bifidobacteriaceae, whereas it had a negative relationship with Streptococcacea and Clostridiaceae. Human milk IL-8 was positively associated with the phylum Bacteroidetes, and negatively associated with Proteobacteria. At the genus level, human milk IL-8 exhibited a positive relationship with Bacteroides, whereas human milk isoleucine had a negative relationship with Bacteroides and Ruminococcus. Pregravid BMI and sex effects were observed. CONCLUSIONS IL-8 in human milk could potentially prepare the infant's immune system to respond effectively to various microorganisms, potentially promoting the growth of beneficial gut bacteria and protecting against pathogens.
Collapse
Affiliation(s)
- Maryam Kebbe
- Faculty of Kinesiology, University of New Brunswick, Fredericton, New Brunswick, Canada
| | - Kartik Shankar
- Department of Pediatrics, University of Colorado, Denver, CO, United States
| | - Leanne M Redman
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| | - Aline Andres
- Arkansas Children's Nutrition Center, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| |
Collapse
|
5
|
Peng LH, Tan Y, Bajinka O. The influence of maternal diet on offspring's gut microbiota in early life. Arch Gynecol Obstet 2024; 309:1183-1190. [PMID: 38057588 DOI: 10.1007/s00404-023-07305-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/12/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND The influence of maternal diet on offspring's health is an area of study that is linked to epigenetics. Maternal diet contributes to determining the health status of offspring and maternally linked mechanisms and is a global health challenge that requires attention. The impact of gut microbiota on host metabolism and offspring health is still not established. OBJECTIVE In this review, we intend to discuss the evidence on the impact of maternal diet and the health of offspring gut microbiota. The paper focuses on the gut microbiome of animal models. It captures the maternal diet and its influence on the offspring's gut microbiota, behavior that is supported by cell experimental results. Both inflammation and immune status of offspring induced by maternal diet are discussed. Finally, this review used predicted biological pathways involved in maternal diet and offspring health, and the influence of maternal diet on gut microbiota and offspring behavior. Obesity, diabetes, asthma and allergies, and neurodegenerative disorders and prospects for maternal diet, and microbiota and offspring health were discussed. CONCLUSION The review was able to gather that a high-fat diet during pregnancy created a long-lasting metabolic signature on the infant's innate immune system, altering inflammation in the offspring microbiota, which predisposed offspring to obesity and metabolic diseases in adulthood.
Collapse
Affiliation(s)
- Li-Hua Peng
- Department of Physiology, Hunan Yongzhou Vocational Technical College, Yongzhou, China
| | - Yurong Tan
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China.
- China-Africa Research Centre of Infectious Diseases, School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China.
| | - Ousman Bajinka
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China.
- China-Africa Research Centre of Infectious Diseases, School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China.
- School of Medicine and Allied Health Sciences, University of The Gambia, Serrekunda, Gambia.
| |
Collapse
|
6
|
Zhang QR, Dong Y, Fan JG. Early-life exposure to gestational diabetes mellitus predisposes offspring to pediatric nonalcoholic fatty liver disease. Hepatobiliary Pancreat Dis Int 2023:S1499-3872(23)00245-X. [PMID: 38195352 DOI: 10.1016/j.hbpd.2023.12.007] [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] [Received: 08/05/2023] [Accepted: 12/28/2023] [Indexed: 01/11/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the prevailing chronic liver disease in the pediatric population due to the global obesity pandemic. Evidence shows that prenatal and postnatal exposure to maternal abnormalities leads to a higher risk of pediatric NAFLD through persistent alterations in developmental programming. Gestational diabetes mellitus (GDM) is a hyperglycemic syndrome which has become the most prevalent complication in pregnant women. An increasing number of both epidemiologic investigations and animal model studies have validated adverse and long-term outcomes in offspring following GDM exposure in utero. Similarly, GDM is considered a crucial risk factor for pediatric NAFLD. This review aimed to summarize currently published studies concerning the inductive roles of GDM in offspring NAFLD development during childhood and adolescence. Dysregulations in hepatic lipid metabolism and gut microbiota in offspring, as well as dysfunctions in the placenta are potential factors in the pathogenesis of GDM-associated pediatric NAFLD. In addition, potentially effective interventions for GDM-associated offspring NAFLD are also discussed in this review. However, most of these therapeutic approaches still require further clinical research for validation.
Collapse
Affiliation(s)
- Qian-Ren Zhang
- Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yan Dong
- Department of Endocrinology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jian-Gao Fan
- Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China.
| |
Collapse
|
7
|
Melo GM, Capucho AM, Sacramento JF, Ponce-de-Leão J, Fernandes MV, Almeida IF, Martins FO, Conde SV. Overnutrition during Pregnancy and Lactation Induces Gender-Dependent Dysmetabolism in the Offspring Accompanied by Heightened Stress and Anxiety. Nutrients 2023; 16:67. [PMID: 38201896 PMCID: PMC10781034 DOI: 10.3390/nu16010067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Maternal obesity and gestational diabetes predispose the next generation to metabolic disturbances. Moreover, the lactation phase also stands as a critical phase for metabolic programming. Nevertheless, the precise mechanisms originating these changes remain unclear. Here, we investigate the consequences of a maternal lipid-rich diet during gestation and lactation and its impact on metabolism and behavior in the offspring. Two experimental groups of Wistar female rats were used: a control group (NC) that was fed a standard diet during the gestation and lactation periods and an overnutrition group that was fed a high-fat diet (HF, 60% lipid-rich) during the same phases. The offspring were analyzed at postnatal days 21 and 28 and at 2 months old (PD21, PD28, and PD60) for their metabolic profiles (weight, fasting glycemia insulin sensitivity, and glucose tolerance) and euthanized for brain collection to evaluate metabolism and inflammation in the hypothalamus, hippocampus, and prefrontal cortex using Western blot markers of synaptic dynamics. At 2 months old, behavioral tests for anxiety, stress, cognition, and food habits were conducted. We observed that the female offspring born from HF mothers exhibited increased weight gain and decreased glucose tolerance that attenuated with age. In the offspring males, weight gain increased at P21 and worsened with age, while glucose tolerance remained unchanged. The offspring of the HF mothers exhibited elevated levels of anxiety and stress during behavioral tests, displaying decreased predisposition for curiosity compared to the NC group. In addition, the offspring from mothers with HF showed increased food consumption and a lower tendency towards food-related aggression. We conclude that exposure to an HF diet during pregnancy and lactation induces dysmetabolism in the offspring and is accompanied by heightened stress and anxiety. There was sexual dimorphism in the metabolic traits but not behavioral phenotypes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Silvia V. Conde
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Rua Camara Pestana, 6, Edificio 2, 1150-082 Lisboa, Portugal; (G.M.M.); (A.M.C.); (J.F.S.); (J.P.-d.-L.); (M.V.F.); (I.F.A.); (F.O.M.)
| |
Collapse
|
8
|
Cecchini L, Barmaz C, Cea MJC, Baeschlin H, Etter J, Netzer S, Bregy L, Marchukov D, Trigo NF, Meier R, Hirschi J, Wyss J, Wick A, Zingg J, Christensen S, Radan AP, Etter A, Müller M, Kaess M, Surbek D, Yilmaz B, Macpherson AJ, Sokollik C, Misselwitz B, Ganal-Vonarburg SC. The Bern Birth Cohort (BeBiCo) to study the development of the infant intestinal microbiota in a high-resource setting in Switzerland: rationale, design, and methods. BMC Pediatr 2023; 23:560. [PMID: 37946167 PMCID: PMC10637001 DOI: 10.1186/s12887-023-04198-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 07/17/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Microbiota composition is fundamental to human health with the intestinal microbiota undergoing critical changes within the first two years of life. The developing intestinal microbiota is shaped by maternal seeding, breast milk and its complex constituents, other nutrients, and the environment. Understanding microbiota-dependent pathologies requires a profound understanding of the early development of the healthy infant microbiota. METHODS Two hundred and fifty healthy pregnant women (≥20 weeks of gestation) from the greater Bern area will be enrolled at Bern University hospital's maternity department. Participants will be followed as mother-baby pairs at delivery, week(s) 1, 2, 6, 10, 14, 24, 36, 48, 96, and at years 5 and 10 after birth. Clinical parameters describing infant growth and development, morbidity, and allergic conditions as well as socio-economic, nutritional, and epidemiological data will be documented. Neuro-developmental outcomes and behavior will be assessed by child behavior checklists at and beyond 2 years of age. Maternal stool, milk, skin and vaginal swabs, infant stool, and skin swabs will be collected at enrolment and at follow-up visits. For the primary outcome, the trajectory of the infant intestinal microbiota will be characterized by 16S and metagenomic sequencing regarding composition, metabolic potential, and stability during the first 2 years of life. Secondary outcomes will assess the cellular and chemical composition of maternal milk, the impact of nutrition and environment on microbiota development, the maternal microbiome transfer at vaginal or caesarean birth and thereafter on the infant, and correlate parameters of microbiota and maternal milk on infant growth, development, health, and mental well-being. DISCUSSION The Bern birth cohort study will provide a detailed description and normal ranges of the trajectory of microbiota maturation in a high-resource setting. These data will be compared to data from low-resource settings such as from the Zimbabwe-College of Health-Sciences-Birth-Cohort study. Prospective bio-sampling and data collection will allow studying the association of the microbiota with common childhood conditions concerning allergies, obesity, neuro-developmental outcomes , and behaviour. Trial registration The trial has been registered at www. CLINICALTRIALS gov , Identifier: NCT04447742.
Collapse
Affiliation(s)
- Luca Cecchini
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Colette Barmaz
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Maria José Coloma Cea
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Hannah Baeschlin
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Julian Etter
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Stefanie Netzer
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Leonie Bregy
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Dmitrij Marchukov
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Nerea Fernandez Trigo
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Rachel Meier
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Jasmin Hirschi
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Jacqueline Wyss
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Andrina Wick
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Joelle Zingg
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Sandro Christensen
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Anda-Petronela Radan
- Department of Obstetrics and Gynaecology, Bern University Hospital, Inselspital, University of Bern, Friedbühlstrasse 19, 3010, Bern, Switzerland
| | - Annina Etter
- Department of Obstetrics and Gynaecology, Bern University Hospital, Inselspital, University of Bern, Friedbühlstrasse 19, 3010, Bern, Switzerland
| | - Martin Müller
- Department of Obstetrics and Gynaecology, Bern University Hospital, Inselspital, University of Bern, Friedbühlstrasse 19, 3010, Bern, Switzerland
| | - Michael Kaess
- University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, Bolligenstrasse 111, Haus A, 3000, Bern, Switzerland
| | - Daniel Surbek
- Department of Obstetrics and Gynaecology, Bern University Hospital, Inselspital, University of Bern, Friedbühlstrasse 19, 3010, Bern, Switzerland
| | - Bahtiyar Yilmaz
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Andrew J Macpherson
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Christiane Sokollik
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital, Inselspital, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland
| | - Benjamin Misselwitz
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland.
| | - Stephanie C Ganal-Vonarburg
- Department for BioMedical Research (DBMR), Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| |
Collapse
|
9
|
Zhang H, Wang Y, Zhang X, Zhang L, Zhao X, Xu Y, Wang P, Liang X, Xue M, Liang H. Maternal Folic Acid Supplementation during Pregnancy Prevents Hepatic Steatosis in Male Offspring of Rat Dams Fed High-Fat Diet, Which Is Associated with the Regulation of Gut Microbiota. Nutrients 2023; 15:4726. [PMID: 38004120 PMCID: PMC10675082 DOI: 10.3390/nu15224726] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Maternal dietary patterns during pregnancy have been demonstrated to impact the structure of the gut microbiota in offspring, altering their susceptibility to diseases. This study is designed to elucidate whether the impact of folic acid supplementation during pregnancy on hepatic steatosis in male offspring of rat dams exposed to a high-fat diet (HFD) is related to gut-liver axis homeostasis. In this study, female rats were administered a HFD and simultaneously supplemented with 5 mg/kg folic acid throughout their pregnancy. Histopathological examination showed that folic acid supplementation effectively ameliorated hepatic lipid accumulation and inflammatory infiltrate in male offspring subjected to a maternal HFD. Maternal folic acid supplementation reduced the abundance of Desulfobacterota and the Firmicutes/Bacteroidota (F/B) ratio in male offspring. The expression of tight junction proteins in the colon was significantly upregulated, and the serum LPS level was significantly reduced. Furthermore, there was a notable reduction in the hepatic expression of the TLR4/NF-κB signaling pathway and subsequent inflammatory mediators. Spearman's correlation analysis revealed significant associations between hepatic inflammation-related indices and several gut microbiota, particularly Desulfobacterota and Lactobacillus. With a reduction in hepatic inflammation, the expression of PPAR-α was upregulated, and the expression of SREBP-1c and its downstream lipid metabolism-related genes was downregulated. In summary, folic acid supplementation during pregnancy modulates gut microbiota and enhances intestinal barrier integrity in male offspring of HFD dams. This helps reduce the LPS leakage and suppress the expression of TLR4/NF-κB pathway in the liver, thereby improving lipid metabolism disorders, and alleviating hepatic steatosis.
Collapse
Affiliation(s)
- Huaqi Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| | - Yutong Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| | - Xinyu Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| | - Li Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| | - Xuenuo Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| | - Yan Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| | - Peng Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| | - Xi Liang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| | - Meilan Xue
- Department of Biochemistry and Molecular Biology, Basic Medical College, Qingdao University, Qingdao 266071, China;
| | - Hui Liang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| |
Collapse
|
10
|
Kian N, Bagheri A, Salmanpour F, Soltani A, Mohajer Z, Samieefar N, Barekatain B, Kelishadi R. Breast feeding, obesity, and asthma association: clinical and molecular views. Clin Mol Allergy 2023; 21:8. [PMID: 37789370 PMCID: PMC10546753 DOI: 10.1186/s12948-023-00189-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 08/28/2023] [Indexed: 10/05/2023] Open
Abstract
Asthma is a chronic condition that affects children worldwide. Accumulating number of studies reported that the prevalence of pediatric obesity and asthma might be altered through breastfeeding. It has been proposed that Leptin, which exists in human milk, is oppositely associated with weight increase in newborns. It may also influence peripheral immune system by promoting TH1 responses and suppressing TH2 cytokines. Leptin influences body weight and immune responses through complex signaling pathways at molecular level. Although previous studies provide explanations for the protective role of breastfeeding against both obesity and asthma, other factors such as duration of breastfeeding, parental, and prenatal factors may confound this relationship which requires further research.
Collapse
Affiliation(s)
- Naghmeh Kian
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Alireza Bagheri
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Fardis Salmanpour
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Student Research Committee, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Afsaneh Soltani
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Zahra Mohajer
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Noosha Samieefar
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Behzad Barekatain
- Division of Neonatology, Department of Pediatrics, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roya Kelishadi
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran.
- USERN Office, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
11
|
Keady MM, Jimenez RR, Bragg M, Wagner JCP, Bornbusch SL, Power ML, Muletz-Wolz CR. Ecoevolutionary processes structure milk microbiomes across the mammalian tree of life. Proc Natl Acad Sci U S A 2023; 120:e2218900120. [PMID: 37399384 PMCID: PMC10334807 DOI: 10.1073/pnas.2218900120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 05/22/2023] [Indexed: 07/05/2023] Open
Abstract
Milk production is an ancient adaptation that unites all mammals. Milk contains a microbiome that can contribute to offspring health and microbial-immunological development. We generated a comprehensive milk microbiome dataset (16S rRNA gene) for the class Mammalia, representing 47 species from all placental superorders, to determine processes structuring milk microbiomes. We show that across Mammalia, milk exposes offspring to maternal bacterial and archaeal symbionts throughout lactation. Deterministic processes of environmental selection accounted for 20% of milk microbiome assembly processes; milk microbiomes were similar from mammals with the same host superorder (Afrotheria, Laurasiathera, Euarchontoglires, and Xenarthra: 6%), environment (marine captive, marine wild, terrestrial captive, and terrestrial wild: 6%), diet (carnivore, omnivore, herbivore, and insectivore: 5%), and milk nutrient content (sugar, fat, and protein: 3%). We found that diet directly and indirectly impacted milk microbiomes, with indirect effects being mediated by milk sugar content. Stochastic processes, such as ecological drift, accounted for 80% of milk microbiome assembly processes, which was high compared to mammalian gut and mammalian skin microbiomes (69% and 45%, respectively). Even amid high stochasticity and indirect effects, our results of direct dietary effects on milk microbiomes provide support for enteromammary trafficking, representing a mechanism by which bacteria are transferred from the mother's gut to mammary gland and then to offspring postnatally. The microbial species present in milk reflect both selective pressures and stochastic processes at the host level, exemplifying various ecological and evolutionary factors acting on milk microbiomes, which, in turn, set the stage for offspring health and development.
Collapse
Affiliation(s)
- Mia M. Keady
- Center for Conservation Genomics, Smithsonian National Zoo and Conservation Biology Institute, Washington, DC20008
- Nelson Institute for Environmental Studies, University of Wisconsin-Madison, Madison, WI53706
| | - Randall R. Jimenez
- Center for Conservation Genomics, Smithsonian National Zoo and Conservation Biology Institute, Washington, DC20008
- Science Team, International Union for Conservation of Nature, 11501San José, Costa Rica
| | - Morgan Bragg
- Center for Conservation Genomics, Smithsonian National Zoo and Conservation Biology Institute, Washington, DC20008
- Department of Environmental Science and Policy, George Mason University, Fairfax, VA22030
| | - Jenna C. P. Wagner
- Nutrition Laboratory and Conservation Ecology Center, Smithsonian National Zoo and Conservation Biology Institute, National Zoological Park, Washington, DC20008
| | - Sally L. Bornbusch
- Center for Conservation Genomics, Smithsonian National Zoo and Conservation Biology Institute, Washington, DC20008
- Department of Nutrition Science, Smithsonian National Zoo and Conservation Biology Institute, Washington, DC20008
| | - Michael L. Power
- Nutrition Laboratory and Conservation Ecology Center, Smithsonian National Zoo and Conservation Biology Institute, National Zoological Park, Washington, DC20008
| | - Carly R. Muletz-Wolz
- Center for Conservation Genomics, Smithsonian National Zoo and Conservation Biology Institute, Washington, DC20008
| |
Collapse
|
12
|
Carson KE, Alvarez J, Mackley J, Travagli RA, Browning KN. Perinatal high-fat diet exposure alters oxytocin and corticotropin releasing factor inputs onto vagal neurocircuits controlling gastric motility. J Physiol 2023; 601:2853-2875. [PMID: 37154244 PMCID: PMC10524104 DOI: 10.1113/jp284726] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/03/2023] [Indexed: 05/10/2023] Open
Abstract
Perinatal high-fat diet (pHFD) exposure alters the development of vagal neurocircuits that control gastrointestinal (GI) motility and reduce stress resiliency in offspring. Descending oxytocin (OXT; prototypical anti-stress peptide) and corticotropin releasing factor (CRF; prototypical stress peptide) inputs from the paraventricular nucleus (PVN) of the hypothalamus to the dorsal motor nucleus of the vagus (DMV) modulate the GI stress response. How these descending inputs, and their associated changes to GI motility and stress responses, are altered following pHFD exposure are, however, unknown. The present study used retrograde neuronal tracing experiments, cerebrospinal fluid extraction, in vivo recordings of gastric tone, motility and gastric emptying rates, and in vitro electrophysiological recordings from brainstem slice preparations to investigate the hypothesis that pHFD alters descending PVN-DMV inputs and dysregulates vagal brain-gut responses to stress. Compared to controls, rats exposed to pHFD had slower gastric emptying rates and did not respond to acute stress with the expected delay in gastric emptying. Neuronal tracing experiments demonstrated that pHFD reduced the number of PVNOXT neurons that project to the DMV, but increased PVNCRF neurons. Both in vitro electrophysiology recordings of DMV neurons and in vivo recordings of gastric motility and tone demonstrated that, following pHFD, PVNCRF -DMV projections were tonically active, and that pharmacological antagonism of brainstem CRF1 receptors restored the appropriate gastric response to brainstem OXT application. These results suggest that pHFD exposure disrupts descending PVN-DMV inputs, leading to a dysregulated vagal brain-gut response to stress. KEY POINTS: Maternal high-fat diet exposure is associated with gastric dysregulation and stress sensitivity in offspring. The present study demonstrates that perinatal high-fat diet exposure downregulates hypothalamic-vagal oxytocin (OXT) inputs but upregulates hypothalamic-vagal corticotropin releasing factor (CRF) inputs. Both in vitro and in vivo studies demonstrated that, following perinatal high-fat diet, CRF receptors were tonically active at NTS-DMV synapses, and that pharmacological antagonism of these receptors restored the appropriate gastric response to OXT. The current study suggests that perinatal high-fat diet exposure disrupts descending PVN-DMV inputs, leading to a dysregulated vagal brain-gut response to stress.
Collapse
Affiliation(s)
- Kaitlin E. Carson
- Department of Neural and Behavioral Sciences, Pennsylvania State College of Medicine, Hershey, PA
| | - Jared Alvarez
- Barrett Honors College, Arizona State University, Tempe, AZ
| | - Jasmine Mackley
- Schreyer Honors College, Pennsylvania State University, State College, PA
| | | | - Kirsteen N. Browning
- Address for correspondence: Kirsteen N. Browning, PhD, Penn State College of Medicine, 500 University Drive, MC H109, Hershey, PA, 17033;
| |
Collapse
|
13
|
Wu AJ, Oken E. Developmental Contributions to Obesity: Nutritional Exposures in the First Thousand Days. Gastroenterol Clin North Am 2023; 52:333-345. [PMID: 37197877 PMCID: PMC10315183 DOI: 10.1016/j.gtc.2023.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Obesity is prevalent and continuing to rise across all age groups, even children. As obesity is challenging to manage and treat, prevention is critical. Here, we highlight nutritional influences during periods of early developmental plasticity, namely the prenatal period and infancy, that have been shown to contribute to the development of obesity into childhood and beyond. We review recent research that examines maternal nutritional factors including dietary patterns and quality, as well as the infant diet, such as complementary foods and beverages, that influence long-term obesity risk. We end with recommendations for clinicians.
Collapse
Affiliation(s)
- Allison J Wu
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Hunnewell Ground, Boston, MA 02115, USA.
| | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, 401 Park Drive, Suite 401E, Boston, MA 02215, USA
| |
Collapse
|
14
|
Sturov NV, Popov SV, Belikov II. Gut Microbiota and the Ways to Correct it in Chronic Kidney Disease. Indian J Nephrol 2023; 33:162-169. [PMID: 37448901 PMCID: PMC10337223 DOI: 10.4103/ijn.ijn_469_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/27/2022] [Accepted: 02/06/2022] [Indexed: 07/15/2023] Open
Abstract
Approximately 13% of the Russian population suffers from chronic kidney disease (CKD). Such a high prevalence of the disease, as well as the complexity and high cost of renal replacement therapy, explain the need for developing and implementing new approaches to treat patients at the pre-dialysis stages. The data collected in recent decades highlight the importance of gut microbiota in the progression of CKD. This review provides information about the microbiota composition in healthy individuals and patients with CKD and discusses the mechanisms of interaction in the intestine-kidney system. The article also presents the specifics of the violation of gut microbiota (GM) and correction thereof in CKD.
Collapse
Affiliation(s)
- Nikolay V. Sturov
- Peoples’ Friendship University of Russia (RUDN University), Department of General Practice, 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation
| | - Sergey V. Popov
- Peoples’ Friendship University of Russia (RUDN University), Department of General Practice, 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation
| | - Igor I. Belikov
- Peoples’ Friendship University of Russia (RUDN University), Department of General Practice, 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation
| |
Collapse
|
15
|
Djekkoun N, Depeint F, Guibourdenche M, Sabbouri HEKE, Corona A, Rhazi L, Gay-Queheillard J, Rouabah L, Biendo M, Al-Salameh A, Lalau JD, Bach V, Khorsi-Cauet H. Perigestational exposure of a combination of a high-fat diet and pesticide impacts the metabolic and microbiotic status of dams and pups; a preventive strategy based on prebiotics. Eur J Nutr 2023; 62:1253-1265. [PMID: 36510012 DOI: 10.1007/s00394-022-03063-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE Metabolic changes during the perinatal period are known to promote obesity and type-2 diabetes in adulthood via perturbation of the microbiota. The risk factors for metabolic disorders include a high-fat diet (HFD) and exposure to pesticide residues. The objective of the present study was to evaluate the effects of perigestational exposure to a HFD and chlorpyrifos (CPF) on glycemia, lipid profiles, and microbial populations in Wistar dams and their female offspring. We also tested a preventive strategy based on treatment with the prebiotic inulin. METHODS From 4 months before gestation to the end of the lactation period, six groups of dams were exposed to either a standard diet, a HFD alone, CPF alone, a combination of a HFD and CPF, and/or inulin supplementation. All female offspring were fed a standard diet from weaning to adulthood. We measured the impacts of these exposures on glycemia, the lipid profile, and the microbiota (composition, metabolite production, and translocation into tissues). RESULTS HFD exposure and CPF + HFD co-exposure induced dysmetabolism and an imbalance in the gut flora in both the dams and the female offspring. Inulin mitigated the impact of exposure to a HFD alone but not that of CPF + HFD co-exposure. CONCLUSION Our results provide a better understanding of the complex interactions between environmental pollutants and diet in early life, including in the context of metabolic diseases.
Collapse
Affiliation(s)
- Narimane Djekkoun
- Laboratoire PeriTox UMR_I 01, Centre Universitaire de Recherche en Santé, CURS-UPJV, University of Picardy Jules Verne, 80054, Amiens cedex 1, France
- Laboratoire de Biologie Cellulaire Et Moléculaire, Mentouri Brothers University of Constantine 1, 2500, Constantine, Algeria
| | - Flore Depeint
- Transformations Et Agro-Ressources ULR7519, Institut Polytechnique UniLaSalle - Université d'Artois, 60026, Beauvais, France
| | - Marion Guibourdenche
- Laboratoire PeriTox UMR_I 01, Centre Universitaire de Recherche en Santé, CURS-UPJV, University of Picardy Jules Verne, 80054, Amiens cedex 1, France
| | - Hiba El Khayat Et Sabbouri
- Laboratoire PeriTox UMR_I 01, Centre Universitaire de Recherche en Santé, CURS-UPJV, University of Picardy Jules Verne, 80054, Amiens cedex 1, France
| | - Aurélie Corona
- Laboratoire PeriTox UMR_I 01, Centre Universitaire de Recherche en Santé, CURS-UPJV, University of Picardy Jules Verne, 80054, Amiens cedex 1, France
| | - Larbi Rhazi
- Transformations Et Agro-Ressources ULR7519, Institut Polytechnique UniLaSalle - Université d'Artois, 60026, Beauvais, France
| | - Jerome Gay-Queheillard
- Laboratoire PeriTox UMR_I 01, Centre Universitaire de Recherche en Santé, CURS-UPJV, University of Picardy Jules Verne, 80054, Amiens cedex 1, France
| | - Leila Rouabah
- Laboratoire de Biologie Cellulaire Et Moléculaire, Mentouri Brothers University of Constantine 1, 2500, Constantine, Algeria
| | - Maurice Biendo
- Laboratoire PeriTox UMR_I 01, Centre Universitaire de Recherche en Santé, CURS-UPJV, University of Picardy Jules Verne, 80054, Amiens cedex 1, France
| | - Abdallah Al-Salameh
- Service Endocrinologie-Diabétologie et Nutrition, CHU Amiens-Picardie, 80000, Amiens, France
| | - Jean-Daniel Lalau
- Service Endocrinologie-Diabétologie et Nutrition, CHU Amiens-Picardie, 80000, Amiens, France
| | - Véronique Bach
- Laboratoire PeriTox UMR_I 01, Centre Universitaire de Recherche en Santé, CURS-UPJV, University of Picardy Jules Verne, 80054, Amiens cedex 1, France
| | - Hafida Khorsi-Cauet
- Laboratoire PeriTox UMR_I 01, Centre Universitaire de Recherche en Santé, CURS-UPJV, University of Picardy Jules Verne, 80054, Amiens cedex 1, France.
| |
Collapse
|
16
|
Radford-Smith DE, Anthony DC. Mechanisms of Maternal Diet-Induced Obesity Affecting the Offspring Brain and Development of Affective Disorders. Metabolites 2023; 13:455. [PMID: 36984895 PMCID: PMC10053489 DOI: 10.3390/metabo13030455] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/16/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Depression and metabolic disease are common disorders that share a bidirectional relationship and continue to increase in prevalence. Maternal diet and maternal behaviour both profoundly influence the developmental trajectory of offspring during the perinatal period. At an epidemiological level, both maternal depression and obesity during pregnancy have been shown to increase the risk of neuropsychiatric disease in the subsequent generation. Considerable progress has been made to understand the mechanisms by which maternal obesity disrupts the developing offspring gut-brain axis, priming offspring for the development of affective disorders. This review outlines such mechanisms in detail, including altered maternal care, the maternal microbiome, inflammation, breast milk composition, and maternal and placental metabolites. Subsequently, offspring may be prone to developing gut-brain interaction disorders with concomitant changes to brain energy metabolism, neurotransmission, and behaviour, alongside gut dysbiosis. The gut microbiome may act as a key modifiable, and therefore treatable, feature of the relationship between maternal obesity and the offspring brain function. Further studies examining the relationship between maternal nutrition, the maternal microbiome and metabolites, and offspring neurodevelopment are warranted to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Daniel E. Radford-Smith
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford OX37JX, UK
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford OX13TA, UK
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX13QT, UK
| | - Daniel C. Anthony
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX13QT, UK
| |
Collapse
|
17
|
Cömert TK, Akpinar F, Erkaya S, Durmaz B, Durmaz R. The effect of pre-pregnancy obesity on gut and meconium microbiome and relationship with fetal growth. J Matern Fetal Neonatal Med 2022; 35:10629-10637. [PMID: 36398501 DOI: 10.1080/14767058.2022.2148098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To investigate the effect of pre-pregnancy obesity on maternal and newborn microbiomes and fetal growth. METHODS Individuals who gained body weight in accordance with the recommendations during pregnancy and normal gestastional age are included in the study and were separated into two groups, normal (n = 20) and obese (n = 20), based on their body mass index (BMI) value of pre-pregnancy. Maternal stool samples collected during the first trimester of pregnancy and meconium samples collected at birth were evaluated using 16S rRNA gene-based microbiome analysis. RESULTS The stool samples of mothers who were obese before pregnancy harbored a higher (59.9 versus 52.3%) relative abundance of Firmicutes and a lower (7.1 versus 4.1%) relative abundance of Proteobacteria than the stool samples of mothers with normal body weight pre-pregnancy. In contrast, in the meconium samples of mothers who were obese pre-pregnancy, compared to those of mothers who had a normal body weight pre-pregnancy, the phylum Firmicutes was less (56.0 versus 69.0%) abundant and Proteobacteria (9.0 versus 8.5%) was more abundant. There was a negative correlation between pre-pregnancy BMI, birth weight, weight/height ratio and alpha diversity indices (Shannon and Chao1). CONCLUSIONS Pre-pregnancy obesity can affect pregnant and newborn gut microbiota, which might related to fetal growth of the newborn.
Collapse
Affiliation(s)
- Tuğba Küçükkasap Cömert
- Department of Nutrition and Dietetics, Gülhane Faculty of Health Sciences, University of Health Sciences Turkey, Ankara, Turkey
| | - Funda Akpinar
- Department of Obstetrics and Gynecology, Ankara Etlik Zübeyde Hanim Health Practice, University of Health Sciences Turkey, Ankara, Turkey
| | - Salim Erkaya
- Department of Obstetrics and Gynecology, Ankara Etlik Zübeyde Hanim Health Practice, University of Health Sciences Turkey, Ankara, Turkey
| | - Bengül Durmaz
- Department of Clinical Microbiology, Faculty of Medicine, Yüksek İhtisas University, Ankara, Turkey
| | - Riza Durmaz
- Department of Clinical Microbiology, Faculty of Medicine, Molecular Microbiology Section, Ankara Yildirim Beyazit University, Ankara, Turkey
| |
Collapse
|
18
|
Rey-Mariño A, Francino MP. Nutrition, Gut Microbiota, and Allergy Development in Infants. Nutrients 2022; 14:nu14204316. [PMID: 36297000 PMCID: PMC9609088 DOI: 10.3390/nu14204316] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/09/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022] Open
Abstract
The process of gut microbiota development in infants is currently being challenged by numerous factors associated with the contemporary lifestyle, including diet. A thorough understanding of all aspects of microbiota development will be necessary for engineering strategies that can modulate it in a beneficial direction. The long-term consequences for human development and health of alterations in the succession pattern that forms the gut microbiota are just beginning to be explored and require much further investigation. Nevertheless, it is clear that gut microbiota development in infancy bears strong associations with the risk for allergic disease. A useful understanding of microbial succession in the gut of infants needs to reveal not only changes in taxonomic composition but also the development of functional capacities through time and how these are related to diet and various environmental factors. Metagenomic and metatranscriptomic studies have started to produce insights into the trends of functional repertoire and gene expression change within the first year after birth. This understanding is critical as during this period the most substantial development of the gut microbiota takes place and the relations between gut microbes and host immunity are established. However, further research needs to focus on the impact of diet on these changes and on how diet can be used to counteract the challenges posed by modern lifestyles to microbiota development and reduce the risk of allergic disease.
Collapse
Affiliation(s)
- Alejandra Rey-Mariño
- Genomics and Health Department, Foundation for the Promotion of Health and Biomedical Research of the Valencia Region (FISABIO), 46020 València, Spain
| | - M. Pilar Francino
- Genomics and Health Department, Foundation for the Promotion of Health and Biomedical Research of the Valencia Region (FISABIO), 46020 València, Spain
- CIBER en Epidemiología y Salud Pública (CIBERESP), 28001 Madrid, Spain
- Correspondence:
| |
Collapse
|
19
|
Hypertension and renal disease programming: focus on the early postnatal period. Clin Sci (Lond) 2022; 136:1303-1339. [PMID: 36073779 DOI: 10.1042/cs20220293] [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: 05/09/2022] [Revised: 08/18/2022] [Accepted: 08/25/2022] [Indexed: 11/17/2022]
Abstract
The developmental origin of hypertension and renal disease is a concept highly supported by strong evidence coming from both human and animal studies. During development there are periods in which the organs are more vulnerable to stressors. Such periods of susceptibility are also called 'sensitive windows of exposure'. It was shown that as earlier an adverse event occurs; the greater are the consequences for health impairment. However, evidence show that the postnatal period is also quite important for hypertension and renal disease programming, especially in rodents because they complete nephrogenesis postnatally, and it is also important during preterm human birth. Considering that the developing kidney is vulnerable to early-life stressors, renal programming is a key element in the developmental programming of hypertension and renal disease. The purpose of this review is to highlight the great number of studies, most of them performed in animal models, showing the broad range of stressors involved in hypertension and renal disease programming, with a particular focus on the stressors that occur during the early postnatal period. These stressors mainly include undernutrition or specific nutritional deficits, chronic behavioral stress, exposure to environmental chemicals, and pharmacological treatments that affect some important factors involved in renal physiology. We also discuss the common molecular mechanisms that are activated by the mentioned stressors and that promote the appearance of these adult diseases, with a brief description on some reprogramming strategies, which is a relatively new and promising field to treat or to prevent these diseases.
Collapse
|
20
|
Maternal and infant NR3C1 and SLC6A4 epigenetic signatures of the COVID-19 pandemic lockdown: when timing matters. Transl Psychiatry 2022; 12:386. [PMID: 36114180 PMCID: PMC9481531 DOI: 10.1038/s41398-022-02160-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/02/2022] [Accepted: 09/07/2022] [Indexed: 11/27/2022] Open
Abstract
Stress exposure during pregnancy is critically linked with maternal mental health and child development. The effects might involve altered patterns of DNA methylation in specific stress-related genes (i.e., glucocorticoid receptor gene, NR3C1, and serotonin transporter gene, SLC6A4) and might be moderated by the gestational timing of stress exposure. In this study, we report on NR3C1 and SLC6A4 methylation status in Italian mothers and infants who were exposed to the COVID-19 pandemic lockdown during different trimesters of pregnancy. From May 2020 to February 2021, 283 mother-infant dyads were enrolled at delivery. Within 24 h from delivery, buccal cells were collected to assess NR3C1 (44 CpG sites) and SLC6A4 (13 CpG sites) methylation status. Principal component (PC) analyses were used to reduce methylation data dimension to one PC per maternal and infant gene methylation. Mother-infant dyads were split into three groups based on the pregnancy trimester (first, second, third), during which they were exposed to the COVID-19 lockdown. Mothers and infants who were exposed to the lockdown during the first trimester of pregnancy had lower NR3C1 and SLC6A4 methylation when compared to counterparts exposed during the second or third trimesters. The effect remained significant after controlling for confounders. Women who were pregnant during the pandemic and their infants might present altered epigenetic biomarkers of stress-related genes. As these epigenetic marks have been previously linked with a heightened risk of maternal psychiatric problems and less-than-optimal child development, mothers and infants should be adequately monitored for psychological health during and after the pandemic.
Collapse
|
21
|
Carter SA, Lin JC, Chow T, Yu X, Rahman MM, Martinez MP, Feldman K, Eckel SP, Chen JC, Chen Z, Levitt P, Lurmann FW, McConnell R, Xiang AH. Maternal obesity, diabetes, preeclampsia, and asthma during pregnancy and likelihood of autism spectrum disorder with gastrointestinal disturbances in offspring. AUTISM : THE INTERNATIONAL JOURNAL OF RESEARCH AND PRACTICE 2022; 27:916-926. [PMID: 36062479 PMCID: PMC9984567 DOI: 10.1177/13623613221118430] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
LAY ABSTRACT Autism spectrum disorder is heterogeneous and often accompanied by co-occurring conditions. Previous studies have shown that maternal health conditions during pregnancy including obesity, diabetes, preeclampsia, and asthma were associated with increased likelihood of autism. However, little has been done examining the likelihood associated with autism with co-occurring conditions. This study assessed these maternal health conditions in relationship to autism and gastrointestinal disturbances, a common co-occurring condition in children diagnosed with autism. Data included 308,536 mother-child pairs from one integrated health care system with comprehensive electronic medical records. Among the study cohort, 5,131 (1.7%) children had a diagnosis of autism by age 5. Gastrointestinal disturbances were present in 35.4% of children diagnosed with autism and 25.1% of children without autism diagnoses. Our results showed that each of the four maternal health conditions during pregnancy was associated with increased likelihood of gastrointestinal disturbances, autism without gastrointestinal disturbances, and autism with gastrointestinal disturbances. For all four maternal health conditions, the association was greatest for likelihood of autism with gastrointestinal disturbances. Given that children diagnosed with autism are more likely to have gastrointestinal disturbances and over 80% of gastrointestinal disturbances in this cohort were diagnosed prior to autism diagnosis, this study suggests that there may be common biological pathways between autism and gastrointestinal disturbances impacted by these maternal exposures. Future studies are warranted to assess associations between different exposures and autism with other co-occurring conditions to increase our understanding of autism heterogeneity.
Collapse
Affiliation(s)
| | - Jane C Lin
- Kaiser Permanente Southern California, USA
| | - Ting Chow
- Kaiser Permanente Southern California, USA
| | - Xin Yu
- University of Southern California, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Dai X, Wang C, Guo Z, Li Y, Liu T, Jin G, Wang S, Wang B, Jiang K, Cao H. Maternal sucralose exposure induces Paneth cell defects and exacerbates gut dysbiosis of progeny mice. Food Funct 2021; 12:12634-12646. [PMID: 34821899 DOI: 10.1039/d1fo02921e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Research has shown that maternal sucralose (MS) exposure alters the gut microbiota of offspring at weaning and predisposes the offspring to developing obesity, non-alcoholic fatty liver disease and metabolic syndrome later in life. However, the underlying mechanism remains unclear. Paneth cells are thought to critically influence the gut microbiota. This study aimed to investigate whether MS exposure induced Paneth cell defects and exacerbated gut dysbiosis of offspring. Female C57BL/6 mice were divided into the MS and control (water) groups during pregnancy and lactation. Progeny mice were fed a normal sucralose-free diet after weaning until adulthood. MS inhibited intestinal development and increased the expression of proinflammatory cytokines in the small intestines of 3-week-old progeny mice. MS increased the proportions of abnormal granule secretion by Paneth cells. The number of Paneth cells and mRNA expression of AMPs such as cryptdins and lysozyme were reduced in the MS group. MS disturbed the gut microbiota composition and diversity in the 3-week-old offspring mice. The relative abundances of pro-inflammatory bacteria, such as Desulfovibrionales, Helicobacter, Pasteurellales and Campylobacterales were significantly increased in the MS group, while anti-inflammatory bacteria, including Clostridium XI, were decreased. This dysbiosis continued into adulthood. These findings showed that MS exposure induced Paneth cell defects and exacerbated gut dysbiosis in offspring mice. Sucralose should be consumed with caution, especially during pregnancy and in early life.
Collapse
Affiliation(s)
- Xin Dai
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Chen Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Zixuan Guo
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Yun Li
- Department of Pharmacy, General Hospital, Tianjin Medical University, Tianjin, China
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Ge Jin
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Sinan Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Kui Jiang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| |
Collapse
|
23
|
Hill L, Sharma R, Hart L, Popov J, Moshkovich M, Pai N. The neonatal microbiome in utero and beyond: perinatal influences and long-term impacts. J LAB MED 2021. [DOI: 10.1515/labmed-2021-0131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
The neonatal microbiome offers a valuable model for studying the origins of human health and disease. As the field of metagenomics expands, we also increase our understanding of early life influences on its development. In this review we will describe common techniques used to define and measure the microbiome. We will review in utero influences, normal perinatal development, and known risk factors for abnormal neonatal microbiome development. Finally, we will summarize current evidence that links early life microbial impacts on the development of chronic inflammatory diseases, obesity, and atopy.
Collapse
Affiliation(s)
- Lee Hill
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
- Department of Human Biology, Division of Exercise Science and Sports Medicine , University of Cape Town , Cape Town , South Africa
| | - Ruchika Sharma
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
- McMaster University , Hamilton , Canada
| | - Lara Hart
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
| | - Jelena Popov
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
- University College Cork, College of Medicine and Health , Cork , Ireland
| | - Michal Moshkovich
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
- Faculty of Health Sciences , McMaster University , Hamilton , Canada
| | - Nikhil Pai
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
- Farncombe Family Digestive Health Research Institute , McMaster University , Hamilton , Canada
| |
Collapse
|
24
|
Conta G, Del Chierico F, Reddel S, Marini F, Sciubba F, Capuani G, Tomassini A, Di Cocco ME, Laforgia N, Baldassarre ME, Putignani L, Miccheli A. Longitudinal Multi-Omics Study of a Mother-Infant Dyad from Breastfeeding to Weaning: An Individualized Approach to Understand the Interactions Among Diet, Fecal Metabolome and Microbiota Composition. Front Mol Biosci 2021; 8:688440. [PMID: 34671642 PMCID: PMC8520934 DOI: 10.3389/fmolb.2021.688440] [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: 03/30/2021] [Accepted: 09/20/2021] [Indexed: 01/02/2023] Open
Abstract
The development of the human gut microbiota is characterized by a dynamic sequence of events from birth to adulthood, which make the gut microbiota unique for everyone. Its composition and metabolism may play a critical role in the intestinal homeostasis and health. We propose a study on a single mother-infant dyad to follow the dynamics of an infant fecal microbiota and metabolome changes in relation to breast milk composition during the lactation period and evaluate the changes induced by introduction of complementary food during the weaning period. Nuclear Magnetic Resonance (NMR)-based metabolomics was performed on breast milk and, together with 16S RNA targeted-metagenomics analysis, also on infant stool samples of a mother-infant dyad collected over a period running from the exclusive breastfeeding diet to weaning. Breast milk samples and neonatal stool samples were collected from the 4th to the 10th month of life. Both specimens were collected from day 103 to day 175, while from day 219-268 only stool samples were examined. An exploratory and a predictive analysis were carried out by means of Common component and specific weight analysis and multi-block partial least squares discriminant analysis, respectively. Stools collected during breastfeeding and during a mixed fruit/breastfeeding diet were characterized by high levels of fucosyl-oligosaccharides and glycolysis intermediates, including succinate and formate. The transition to a semi-solid food diet was characterized by several changes in fecal parameters: increase in short-chain fatty acids (SCFAs) levels, including acetate, propionate and butyrate, dissapearance of HMOs and the shift in the community composition, mainly occurring within the Firmicutes phylum. The variations in the fecal metabolome reflected the infant's diet transition, while the composition of the microbiota followed a more complex and still unstable behavior.
Collapse
Affiliation(s)
- Giorgia Conta
- Department of Chemistry, Sapienza University of Rome, Rome, Italy.,NMR-Based Metabolomics Laboratory of Sapienza (NMLab), Sapienza University of Rome, Rome, Italy
| | - Federica Del Chierico
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sofia Reddel
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Federico Marini
- Department of Chemistry, Sapienza University of Rome, Rome, Italy.,NMR-Based Metabolomics Laboratory of Sapienza (NMLab), Sapienza University of Rome, Rome, Italy
| | - Fabio Sciubba
- Department of Chemistry, Sapienza University of Rome, Rome, Italy.,NMR-Based Metabolomics Laboratory of Sapienza (NMLab), Sapienza University of Rome, Rome, Italy
| | - Giorgio Capuani
- Department of Chemistry, Sapienza University of Rome, Rome, Italy.,NMR-Based Metabolomics Laboratory of Sapienza (NMLab), Sapienza University of Rome, Rome, Italy
| | | | | | - Nicola Laforgia
- Department of Biomedical Science and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | | | - Lorenza Putignani
- Department of Diagnostic and Laboratory Medicine, Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Alfredo Miccheli
- NMR-Based Metabolomics Laboratory of Sapienza (NMLab), Sapienza University of Rome, Rome, Italy.,Department of Environmental Biology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
25
|
Daliry A, Pereira ENGDS. Role of Maternal Microbiota and Nutrition in Early-Life Neurodevelopmental Disorders. Nutrients 2021; 13:3533. [PMID: 34684534 PMCID: PMC8540774 DOI: 10.3390/nu13103533] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/14/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023] Open
Abstract
The rise in the prevalence of obesity and other related metabolic diseases has been paralleled by an increase in the frequency of neurodevelopmental problems, which has raised the likelihood of a link between these two phenomena. In this scenario, maternal microbiota is a possible linking mechanistic pathway. According to the "Developmental Origins of Health and Disease" paradigm, environmental exposures (in utero and early life) can permanently alter the body's structure, physiology, and metabolism, increasing illness risk and/or speeding up disease progression in offspring, adults, and even generations. Nutritional exposure during early developmental stages may induce susceptibility to the later development of human diseases via interactions in the microbiome, including alterations in brain function and behavior of offspring, as explained by the gut-brain axis theory. This review provides an overview of the implications of maternal nutrition on neurodevelopmental disorders and the establishment and maturation of gut microbiota in the offspring.
Collapse
Affiliation(s)
- Anissa Daliry
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil;
| | | |
Collapse
|
26
|
Gawlik A, Salonen A, Jian C, Yanover C, Antosz A, Shmoish M, Wasniewska M, Bereket A, Wudy SA, Hartmann MF, Thivel D, Matusik P, Weghuber D, Hochberg Z. Personalized approach to childhood obesity: Lessons from gut microbiota and omics studies. Narrative review and insights from the 29th European childhood obesity congress. Pediatr Obes 2021; 16:e12835. [PMID: 34296826 DOI: 10.1111/ijpo.12835] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 06/20/2021] [Accepted: 07/05/2021] [Indexed: 12/19/2022]
Abstract
The traditional approach to childhood obesity prevention and treatment should fit most patients, but misdiagnosis and treatment failure could be observed in some cases that lie away from average as part of individual variation or misclassification. Here, we reflect on the contributions that high-throughput technologies such as next-generation sequencing, mass spectrometry-based metabolomics and microbiome analysis make towards a personalized medicine approach to childhood obesity. We hypothesize that diagnosing a child as someone with obesity captures only part of the phenotype; and that metabolomics, genomics, transcriptomics and analyses of the gut microbiome, could add precision to the term "obese," providing novel corresponding biomarkers. Identifying a cluster -omic signature in a given child can thus facilitate the development of personalized prognostic, diagnostic, and therapeutic approaches. It can also be applied to the monitoring of symptoms/signs evolution, treatment choices and efficacy, predisposition to drug-related side effects and potential relapse. This article is a narrative review of the literature and summary of the main observations, conclusions and perspectives raised during the annual meeting of the European Childhood Obesity Group. Authors discuss some recent advances and future perspectives on utilizing a systems approach to understanding and managing childhood obesity in the context of the existing omics data.
Collapse
Affiliation(s)
- Aneta Gawlik
- Department of Paediatrics and Paediatric Endocrinology, Faculty of Medical Sciences, Medical University of Silesia, Katowice, Poland
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ching Jian
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Chen Yanover
- Healthcare Informatics, IBM Research-Haifa, Haifa, Israel
| | - Aleksandra Antosz
- Department of Paediatrics and Paediatric Endocrinology, Faculty of Medical Sciences, Medical University of Silesia, Katowice, Poland
| | - Michael Shmoish
- Bioinformatics Knowledge Unit, The Lokey Centre, Technion - Israel Institute of Technology, Haifa, Israel
| | - Malgorzata Wasniewska
- Department of Human Pathology in Adulthood and Childhood, University of Messina, Messina, Italy
| | - Abdullah Bereket
- School of Medicine, Department of Paediatric Endocrinology, Marmara University, Istanbul, Turkey
| | - Stefan A Wudy
- Steroid Research & Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics, Division of Paediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus-Liebig-University, Giessen, Germany
| | - Michaela F Hartmann
- Steroid Research & Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics, Division of Paediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus-Liebig-University, Giessen, Germany
| | - David Thivel
- University Clermont Auvergne, UFR Medicine, Clermont-Ferrand, France
| | - Pawel Matusik
- Department of Paediatrics and Paediatric Endocrinology, Faculty of Medical Sciences, Medical University of Silesia, Katowice, Poland
| | - Daniel Weghuber
- Department of Paediatrics, Paracelsus Medical University, Salzburg, Austria
| | - Ze'ev Hochberg
- Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
27
|
Li J, Wan Y, Zheng Z, Zhang H, Li Y, Guo X, Li K, Li D. Maternal n-3 polyunsaturated fatty acids restructure gut microbiota of offspring mice and decrease their susceptibility to mammary gland cancer. Food Funct 2021; 12:8154-8168. [PMID: 34291263 DOI: 10.1039/d1fo00906k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Our previous studies have revealed that a maternal diet rich in n-3 polyunsaturated fatty acids (PUFAs) is associated with decreased mammary cancer risk in offspring. However, the underlying mechanism remains unclear. The present study aimed to investigate the possible mechanism by which maternal n-3 PUFAs decrease the mammary cancer risk of offspring in terms of gut microbiota. C57BL/6 pregnant mice were fed a control standard chow (CON), fish oil supplemented diet (n-3 Sup-FO), flaxseed oil supplemented diet (n-3 Sup-FSO) or n-3 PUFA deficient diet (n-3 Def) (n = 10) throughout gestation and lactation. After weaning, all offspring were fed a AIN-93G diet. The tumor incidence and volume were significantly increased in n-3 Def offspring compared with the other groups. Maternal n-3 PUFA supplementation resulted in a significantly increased α-diversity of the gut microbiota in n-3 Sup-FO and n-3 Sup-FSO offspring compared with that in n-3 Def offspring. The relative abundances of Akkermansia, Lactobacillus and Mucispirillum observed in adult offspring of both the n-3 Sup-FO and n-3 Sup-FSO groups were higher than those observed in the control group, whereas the maternal n-3 Def diet was associated with decreased abundances of Lactobacillus, Bifidobacterium and Barnesiella in 7-week-old offspring. The levels of the pro-inflammatory factors IL-1β, IL-6 and TNF-α were significantly lower in n-3 PUFA supplemented offspring than in n-3 Def offspring. In addition, the abundance of Mucispirillum was positively associated with the concentration of the anti-inflammatory factor IL-10, whereas the abundances of Bifidobacterium and Akkermansia were negatively associated with IL-1β and IL-6, respectively. Based on the bacterial composition of the gut microbiota, metabolites were predicted and the results showed that arachidonic acid metabolism and the MAPK signaling pathways were more enriched, while the butyric acid metabolic pathway was less enriched in offspring of the n-3 Def group than in those of the other three groups. Our findings suggest that decreased pro-inflammatory factors and changed gut microbiota are associated with the protective effects of maternal n-3 PUFAs against offspring's mammary tumorigenesis.
Collapse
Affiliation(s)
- Jiaomei Li
- Institute of Nutrition and Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Riederer M, Schweighofer N, Trajanoski S, Stelzer C, Zehentner M, Fuchs-Neuhold B, Kashofer K, Mayr JA, Hörmann-Wallner M, Holasek S, van der Kleyn M. Free threonine in human breast milk is related to infant intestinal microbiota composition. Amino Acids 2021; 54:365-383. [PMID: 34477981 PMCID: PMC8948153 DOI: 10.1007/s00726-021-03057-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/26/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Accumulating evidence indicates that free amino acids (FAA) might be bioactive compounds with potential immunomodulatory capabilities. However, the FAA composition in human milk is still poorly characterized with respect to its correlation to maternal serum levels and its physiological significance for the infant. Studies addressing the relation of human milk FAA to the infants' intestinal microbiota are still missing. METHODS As part of a pilot study, maternal serum and breast milk FAA concentrations as well as infant intestinal microbiota (16S rRNA) were determined 2 months after birth. The study cohort consisted of 41 healthy mothers and their term delivered, healthy infants with normal birthweight. The relationship between maternal serum and milk FAA was determined by correlation analyses. Associations between (highly correlated) milk FAA and infant intestinal beta diversity were tested using PERMANOVA, LefSe and multivariate regression models adjusted for common confounders. RESULTS Seven breast milk FAA correlated significantly with serum concentrations. One of these, threonine showed a negative association with abundance of members of the class Gammaproteobacteria (R2adj = 17.1%, p = 0.006; β= - 0.441). In addition, on the level of families and genera, threonine explained 23.2% of variation of the relative abundance of Enterobacteriaceae (R2adj; p = 0.001; β = - 0.504) and 11.1% of variability in the abundance of Escherichia/Shigella (R2adj, p = 0.025; β = - 0.368), when adjusted for confounders. CONCLUSION Our study is the first to suggest potential interactions between breast milk FAA and infant gut microbiota composition during early lactation. The results might be indicative of a potential protective role of threonine against members of the Enterobacteriaceae family in breast-fed infants. Still, results are based on correlation analyses and larger cohorts are needed to support the findings and elucidate possible underlying mechanisms to assess the complex interplay between breast milk FAA and infant intestinal microbiota in detail.
Collapse
Affiliation(s)
- Monika Riederer
- Institute of Biomedical Science, University of Applied Sciences JOANNEUM, Graz, Austria.
| | | | - Slave Trajanoski
- Core Facility Computational Bioanalytics, Center for Medical Research (ZMF), Medical University of Graz, Graz, Austria
| | - Claudia Stelzer
- Institute of Biomedical Science, University of Applied Sciences JOANNEUM, Graz, Austria
| | - Miriam Zehentner
- Institute of Biomedical Science, University of Applied Sciences JOANNEUM, Graz, Austria
| | - Bianca Fuchs-Neuhold
- Institute of Dietetics and Nutrition, Health Perception Lab, University of Applied Sciences JOANNEUM, Graz, Austria
| | - Karl Kashofer
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Auenbruggerpl. 2, 8036, Graz, Austria
| | - Johannes A Mayr
- Department of Pediatrics, Salzburger Landeskliniken and Paracelsus Medical University, Salzburg, Austria
| | - Marlies Hörmann-Wallner
- Institute of Dietetics and Nutrition, Health Perception Lab, University of Applied Sciences JOANNEUM, Graz, Austria
| | - Sandra Holasek
- Department of Pathophysiology, Medical University Graz, Graz, Austria
| | | |
Collapse
|
29
|
Lee J, Garcia V, Nambiar SM, Jiang H, Dai G. Activation of Proneuronal Transcription Factor Ascl1 in Maternal Liver Ensures a Healthy Pregnancy. Cell Mol Gastroenterol Hepatol 2021; 13:35-55. [PMID: 34438112 PMCID: PMC8600092 DOI: 10.1016/j.jcmgh.2021.08.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 08/08/2021] [Accepted: 08/10/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Maternal liver shows robust adaptations to pregnancy to accommodate the metabolic needs of the developing and growing placenta and fetus by largely unknown mechanisms. We found that Ascl1, a gene encoding a basic helix-loop-helix transcription factor essential for neuronal development, is highly activated in maternal hepatocytes during the second half of gestation in mice. METHODS To investigate whether and how Ascl1 plays a pregnancy-dependent role, we deleted the Ascl1 gene specifically in maternal hepatocytes from midgestation until term. RESULTS As a result, we identified multiple Ascl1-dependent phenotypes. Maternal livers lacking Ascl1 showed aberrant hepatocyte structure, increased hepatocyte proliferation, enlarged hepatocyte size, reduced albumin production, and increased release of liver enzymes, indicating maternal liver dysfunction. Simultaneously, maternal pancreas and spleen and the placenta showed marked overgrowth; and the maternal ceca microbiome showed alterations in relative abundance of several bacterial subpopulations. Moreover, litters born from maternal hepatic Ascl1-deficient dams experienced abnormal postnatal growth after weaning, implying an adverse pregnancy outcome. Mechanistically, we found that maternal hepatocytes deficient for Ascl1 showed robust activation of insulin-like growth factor 2 expression, which may contribute to the Ascl1-dependent phenotypes widespread in maternal and uteroplacental compartments. CONCLUSIONS In summary, we show that maternal liver, via activating Ascl1 expression, modulates the adaptations of maternal organs and the growth of the placenta to maintain a healthy pregnancy. Our studies show that Ascl1 is a novel and critical regulator of the physiology of pregnancy.
Collapse
Affiliation(s)
- Joonyong Lee
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| | - Veronica Garcia
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| | - Shashank M Nambiar
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| | - Huaizhou Jiang
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana; School of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China.
| | - Guoli Dai
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana.
| |
Collapse
|
30
|
Chleilat F, Schick A, Deleemans JM, Ma K, Alukic E, Wong J, Noye Tuplin EW, Nettleton JE, Reimer RA. Paternal high protein diet modulates body composition, insulin sensitivity, epigenetics, and gut microbiota intergenerationally in rats. FASEB J 2021; 35:e21847. [PMID: 34405464 DOI: 10.1096/fj.202100198rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/19/2022]
Abstract
Mounting evidence demonstrates that paternal diet programs offspring metabolism. However, the contribution of a pre-conception paternal high protein (HP) diet to offspring metabolism, gut microbiota, and epigenetic changes remains unclear. Here we show that paternal HP intake in Sprague Dawley rats programs protective metabolic outcomes in offspring. Compared to paternal high fat/sucrose (HF/S), HP diet improved body composition and insulin sensitivity and improved circulating satiety hormones and cecal short-chain fatty acids compared to HF/S and control diet (P < .05). Further, using 16S rRNA gene sequencing to assess gut microbial composition, we observed increased alpha diversity, distinct bacterial clustering, and increased abundance of Bifidobacterium, Akkermansia, Bacteroides, and Marvinbryantia in HP fathers and/or male and female adult offspring. At the epigenetic level, DNMT1and 3b expression was altered intergenerationally. Our study identifies paternal HP diet as a modulator of gut microbial composition, epigenetic markers, and metabolic function intergenerationally.
Collapse
Affiliation(s)
- Faye Chleilat
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Alana Schick
- International Microbiome Centre, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Julie M Deleemans
- Division of Medical Science and Psychosocial Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kyle Ma
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Erna Alukic
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Jolene Wong
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | | | - Jodi E Nettleton
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Raylene A Reimer
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada.,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
31
|
Zhang Z, Xue C, Ju M, Guo J, Wang M, Yi S, Yi X. Maternal Gut Dysbiosis Alters Offspring Microbiota and Social Interactions. Microorganisms 2021; 9:microorganisms9081742. [PMID: 34442821 PMCID: PMC8401725 DOI: 10.3390/microorganisms9081742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 12/22/2022] Open
Abstract
Increasing application of antibiotics changes the gut microbiota composition, leading to dysbiosis of the gut microbiota. Although growing evidence suggests the potential role of gut dysbiosis as the cause of neurodevelopmental disorders and behavioral defects, a broad gap of knowledge remains to be narrowed to better understand the exact mechanisms by which maternal gut dysbiosis alters microbiota development and social interactions of offspring. Here, we showed that maternal gut dysbiosis during gestation is a critical determinant of gut microbiota and social interactions off mouse offspring. Gut microbiota of 2-week-old offspring showed significant changes in response to maternal antibiotic treatment. We even detected distinct effects of maternal oral antibiotics on gut microbiota of 14-week-old offspring. Compared to controls, offspring born to antibiotics-treated mothers displayed reduction in sociability and preference for social novelty, suggesting that the altered offspring social behavior was closely linked to dysbiosis of maternal gut microbiota. Our study opens the possibility to better understand the mechanism of how maternal gut microbiota vertically impairs social interactions of offspring in animal models, providing support to the maternal gut microbiota as a potential mediator between offspring microbiota and behaviors.
Collapse
Affiliation(s)
- Zihan Zhang
- College of Life Sciences, Qufu Normal University, Qufu 273165, China; (Z.Z.); (C.X.); (M.J.); (J.G.); (M.W.)
| | - Chao Xue
- College of Life Sciences, Qufu Normal University, Qufu 273165, China; (Z.Z.); (C.X.); (M.J.); (J.G.); (M.W.)
| | - Mengyao Ju
- College of Life Sciences, Qufu Normal University, Qufu 273165, China; (Z.Z.); (C.X.); (M.J.); (J.G.); (M.W.)
| | - Jiawei Guo
- College of Life Sciences, Qufu Normal University, Qufu 273165, China; (Z.Z.); (C.X.); (M.J.); (J.G.); (M.W.)
| | - Minghui Wang
- College of Life Sciences, Qufu Normal University, Qufu 273165, China; (Z.Z.); (C.X.); (M.J.); (J.G.); (M.W.)
| | - Sijie Yi
- College of Life and Environmental Sciences, University of Exeter, Exeter EX4 4QD, UK
- Correspondence: (S.Y.); (X.Y.)
| | - Xianfeng Yi
- College of Life Sciences, Qufu Normal University, Qufu 273165, China; (Z.Z.); (C.X.); (M.J.); (J.G.); (M.W.)
- Correspondence: (S.Y.); (X.Y.)
| |
Collapse
|
32
|
Jeong S. Early Life Events and Development of Gut Microbiota in Infancy. THE KOREAN JOURNAL OF GASTROENTEROLOGY 2021. [DOI: 10.4166/kjg.2021.408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- SuJin Jeong
- Division of Gastroenterology and Nutrition of Pediatrics, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| |
Collapse
|
33
|
Jian C, Carpén N, Helve O, de Vos WM, Korpela K, Salonen A. Early-life gut microbiota and its connection to metabolic health in children: Perspective on ecological drivers and need for quantitative approach. EBioMedicine 2021; 69:103475. [PMID: 34256346 PMCID: PMC8324810 DOI: 10.1016/j.ebiom.2021.103475] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/18/2021] [Accepted: 06/18/2021] [Indexed: 12/15/2022] Open
Abstract
The colonisation and development of the gut microbiota has been implicated in paediatric metabolic disorders via its powerful effect on host metabolic and immune homeostasis. Here we summarise the evidence from human studies on the early gut microbiota and paediatric overweight and obesity. Manipulation of the early gut microbiota may represent a promising target for countering the burgeoning metabolic disorders in the paediatric population, provided the assembly patterns of microbiota and their health consequences can be decoded. Therefore, in this review, we pay particular attention to the important ecological drivers affecting the community dynamics of the early gut microbiota. We then discuss the knowledge gaps in commonly studied exposures linking the gut microbiota to metabolic disorders, especially regarding maternal factors and antibiotic use. This review also attempts to give directions for future studies aiming to identify predictive and corrective measures for paediatric metabolic disorders based on the gut microbiota. Gut microbiota; Metabolism; Paediatric overweight and obesity; Ecological driver; Dynamics; Infants.
Collapse
Affiliation(s)
- Ching Jian
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Noora Carpén
- Children's Hospital, Pediatric Research Center, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Otto Helve
- Children's Hospital, Pediatric Research Center, Helsinki University Hospital, University of Helsinki, Helsinki, Finland; Finnish Institute for Health and Welfare, Department of Health Security, Helsinki, Finland
| | - Willem M de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Laboratory of Microbiology, Wageningen University & Research, Wageningen, Netherlands
| | - Katri Korpela
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
34
|
Gantenbein KV, Kanaka-Gantenbein C. Mediterranean Diet as an Antioxidant: The Impact on Metabolic Health and Overall Wellbeing. Nutrients 2021; 13:nu13061951. [PMID: 34204057 PMCID: PMC8227318 DOI: 10.3390/nu13061951] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 02/06/2023] Open
Abstract
It has been established, worldwide, that non-communicable diseases such as obesity, diabetes, metabolic syndrome, and cardiovascular events account for a high percentage of morbidity and mortality in contemporary societies. Several modifiable risk factors, such as sedentary activities, sleep deprivation, smoking, and unhealthy dietary habits have contributed to this increase. Healthy nutrition in terms of adherence to the Mediterranean diet (MD), rich in fruits, legumes, vegetables, olive oil, herbs, spices, and high fiber intake may contribute to the decrease in this pandemic. The beneficial effects of the MD can be mainly attributed to its numerous components rich in anti-inflammatory and antioxidant properties. Moreover, the MD may further contribute to the improvement of reproductive health, modify the risk for neurodegenerative diseases, and protect against depression and psychosocial maladjustment. There is also evidence highlighting the impact of healthy nutrition in female people on the composition of the gut microbiota and future metabolic and overall health of their offspring. It is therefore important to highlight the beneficial effects of the MD on metabolic, reproductive, and mental health, while shaping the overall health of future generations. The beneficial effects of MD can be further enhanced by increased physical activity in the context of a well-balanced healthy lifestyle.
Collapse
|
35
|
Alabduljabbar S, Zaidan SA, Lakshmanan AP, Terranegra A. Personalized Nutrition Approach in Pregnancy and Early Life to Tackle Childhood and Adult Non-Communicable Diseases. Life (Basel) 2021; 11:life11060467. [PMID: 34073649 PMCID: PMC8224671 DOI: 10.3390/life11060467] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/05/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023] Open
Abstract
The development of childhood and adult non-communicable diseases (NCD) is associated with environmental factors, starting from intrauterine life. A new theory finds the roots of epigenetic programming in parental gametogenesis, continuing during embryo development, fetal life, and finally in post-natal life. Maternal health status and poor nutrition are widely recognized as implications in the onset of childhood and adult diseases. Early nutrition, particularly breastfeeding, also plays a primary role in affecting the health status of an individual later in life. A poor maternal diet during pregnancy and lack of breastfeeding can cause a nutrient deficiency that affects the gut microbiota, and acts as a cofactor for many pathways, impacting the epigenetic controls and transcription of genes involved in the metabolism, angiogenesis, and other pathways, leading to NCDs in adult life. Both maternal and fetal genetic backgrounds also affect nutrient adsorption and functioning at the cellular level. This review discusses the most recent evidence on maternal nutrition and breastfeeding in the development of NCD, the potentiality of the omics technologies in uncovering the molecular mechanisms underlying it, with the future prospective of applying a personalized nutrition approach to prevent and treat NCD from the beginning of fetal life.
Collapse
|
36
|
Khine WWT, Teo AHT, Loong LWW, Tan JJH, Ang CGH, Ng W, Lee CN, Zhu C, Lau QC, Lee YK. Gut Microbiome of a Multiethnic Community Possessed No Predominant Microbiota. Microorganisms 2021; 9:microorganisms9040702. [PMID: 33805276 PMCID: PMC8065435 DOI: 10.3390/microorganisms9040702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/17/2021] [Accepted: 03/24/2021] [Indexed: 11/16/2022] Open
Abstract
With increasing globalisation, various diets from around the world are readily available in global cities. This study aimed to verify if multiethnic dietary habits destabilised the gut microbiome in response to frequent changes, leading to readily colonisation of exogenous microbes. This may have health implications. We profiled Singapore young adults of different ethnicities for dietary habits, faecal type, gut microbiome and cytokine levels. Subjects were challenged with Lactobacillus casei, and corresponding changes in microbiome and cytokines were evaluated. Here, we found that the majority of young adults had normal stool types (73% Bristol Scale Types 3 and 4) and faecal microbiome categorised into three clusters, irrespective of race and gender. Cluster 1 was dominated by Bacteroides, Cluster 2 by Prevotella, while Cluster 3 showed a marginal increase in Blautia, Ruminococaceae and Ruminococcus, without a predominant microbiota. These youngsters in the three faecal microbiome clusters preferred Western high sugary beverages, Southeast Asian plant-rich diet and Asian/Western diets in rotation, respectively. Multiethnic dietary habits (Cluster 3) led to a gut microbiome without predominant microbiota yet demonstrated colonisation resistance to Lactobacillus. Although Bacteroides and Prevotella are reported to be health-promoting but also risk factors for some illnesses, Singapore-style dietary rotation habits may alleviate Bacteroides and Prevotella associated ill effects. Different immunological outcome was observed during consumption of the lactobacilli among the three microbiome clusters.
Collapse
Affiliation(s)
- Wei Wei Thwe Khine
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore; (W.W.T.K.); (A.H.T.T.)
- Functional Food Forum, Faculty of Medicine, University of Turku, 20014 Turku, Finland
| | - Anna Hui Ting Teo
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore; (W.W.T.K.); (A.H.T.T.)
| | - Lucas Wee Wei Loong
- School of Life Sciences & Chemical Technology, Ngee Ann Polytechnic, 535, Clementi Road, Singapore 599489, Singapore; (L.W.W.L.); (J.J.H.T.); (C.G.H.A.); (W.N.); (C.Z.); (Q.C.L.)
| | - Jarett Jun Hao Tan
- School of Life Sciences & Chemical Technology, Ngee Ann Polytechnic, 535, Clementi Road, Singapore 599489, Singapore; (L.W.W.L.); (J.J.H.T.); (C.G.H.A.); (W.N.); (C.Z.); (Q.C.L.)
| | - Clarabelle Geok Hui Ang
- School of Life Sciences & Chemical Technology, Ngee Ann Polytechnic, 535, Clementi Road, Singapore 599489, Singapore; (L.W.W.L.); (J.J.H.T.); (C.G.H.A.); (W.N.); (C.Z.); (Q.C.L.)
| | - Winnie Ng
- School of Life Sciences & Chemical Technology, Ngee Ann Polytechnic, 535, Clementi Road, Singapore 599489, Singapore; (L.W.W.L.); (J.J.H.T.); (C.G.H.A.); (W.N.); (C.Z.); (Q.C.L.)
| | - Chuen Neng Lee
- Department of Surgery, National University of Hospital, Tower Block, 1E Kent Ridge Road, Singapore 119228, Singapore;
| | - Congju Zhu
- School of Life Sciences & Chemical Technology, Ngee Ann Polytechnic, 535, Clementi Road, Singapore 599489, Singapore; (L.W.W.L.); (J.J.H.T.); (C.G.H.A.); (W.N.); (C.Z.); (Q.C.L.)
| | - Quek Choon Lau
- School of Life Sciences & Chemical Technology, Ngee Ann Polytechnic, 535, Clementi Road, Singapore 599489, Singapore; (L.W.W.L.); (J.J.H.T.); (C.G.H.A.); (W.N.); (C.Z.); (Q.C.L.)
| | - Yuan-Kun Lee
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore; (W.W.T.K.); (A.H.T.T.)
- Department of Surgery, National University of Hospital, Tower Block, 1E Kent Ridge Road, Singapore 119228, Singapore;
- Correspondence:
| |
Collapse
|
37
|
Bayaga CLT, Tanguilig KMN, Aba RPM, Pico MB, Gabriel AA. Culturable micro-organisms in human milk were found to be associated with maternal weight, diet and age during early lactation. J Appl Microbiol 2021; 131:925-937. [PMID: 33336459 DOI: 10.1111/jam.14974] [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: 08/10/2020] [Revised: 12/07/2020] [Accepted: 12/12/2020] [Indexed: 11/26/2022]
Abstract
AIMS This study aimed to evaluate the influence of maternal factors on the total plate count, Staphylococci, Lactobacilli and Bifidobacteria populations in the milk of lactating Filipino women for the first 4 months of lactation period. METHODS AND RESULTS Milk samples (n = 136), 24 h diet recalls and responses from a qualitative food frequency questionnaire (FFQ) were collected from 34 healthy lactating women classified according to their BMI: underweight (n = 7), normal weight (n = 16) and overweight (n = 11). The FFQ was useful in assessing prebiotic and probiotic food items consumed by the participants. Microbial populations were enumerated using culture-plating method, and showed a nonsignificant decreasing trend suggesting their relative stability throughout the first 4 months of lactation. Carbohydrate and fat intakes were associated with TPC, and with both TPC and Staphylococci respectively (P ≤ 0·05); and consumption of root crop is linked with Staphylococci, Lactobacilli and Bifidobacteria (P ≤ 0·05). Interestingly, age was found to be a positive determinant for Bifidobacteria (P = 0·00), whereas being normal- or overweight as negative determinants of Lactobacilli (P = 0·017). Consumption of milk also seems to positively influence both Lactobacilli (P = 0·00) and Bifidobacteria (P = 0·05) counts. CONCLUSIONS Certain populations of culturable micro-organisms were found to be associated with maternal diet, weight classification and age. SIGNIFICANCE AND IMPACT OF THE STUDY This study offered new discoveries in the recently growing endeavor on the role of maternal factors in modulating certain microbial populations in human milk. Ultimately, the findings of this study could provide a basis in crafting lactation policies and guidelines that may help enhance the microbial quality of human milk through adjustments in maternal diet or weight during lactation.
Collapse
Affiliation(s)
- C L T Bayaga
- Breast Milk Laboratory, Department of Food Science and Nutrition, College of Home Economics, University of the Philippines Diliman, Quezon City, Philippines
| | - K M N Tanguilig
- Laboratory of Food Microbiology and Hygiene, Department of Food Science and Nutrition, College of Home Economics, University of the Philippines Diliman, Quezon City, Philippines
| | - R P M Aba
- Laboratory of Food Microbiology and Hygiene, Department of Food Science and Nutrition, College of Home Economics, University of the Philippines Diliman, Quezon City, Philippines
| | - M B Pico
- Breast Milk Laboratory, Department of Food Science and Nutrition, College of Home Economics, University of the Philippines Diliman, Quezon City, Philippines
| | - A A Gabriel
- Laboratory of Food Microbiology and Hygiene, Department of Food Science and Nutrition, College of Home Economics, University of the Philippines Diliman, Quezon City, Philippines
| |
Collapse
|
38
|
Gaukroger CH, Edwards SA, Walshaw J, Nelson A, Adams IP, Stewart CJ, Kyriazakis I. Shifting sows: longitudinal changes in the periparturient faecal microbiota of primiparous and multiparous sows. Animal 2020; 15:100135. [PMID: 33573959 DOI: 10.1016/j.animal.2020.100135] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/29/2020] [Accepted: 11/09/2020] [Indexed: 12/16/2022] Open
Abstract
Knowledge of periparturient longitudinal changes in sow microbiota composition is necessary to fully understand her role in the development of the piglet microbiota, but also to improve gut health and performance of the sow in lactation. Primiparous sows face the challenge of partitioning nutrients to support maternal growth in addition to supporting foetal growth and the demands of lactation. Additional metabolic stress present during the periparturient period may induce changes in the microbiota profile between primiparous and multiparous sows. Using 16S rRNA gene sequencing, the study aimed to characterise the longitudinal changes in the periparturient microbiota and identify differences within the sow microbiota profile associated with parity. Faecal samples from primiparous (n = 13) and multiparous (n = 16) sows were collected at four different time points (day -6, -1, 3 and 8) in relation to farrowing (day 0). Microbiota richness was lowest on day 3 and -1 of the periparturient period (P < 0.05). Microbiota community composition, assessed by weighted and unweighted UniFrac distances, demonstrated longitudinal changes, with day 3 samples clustering away from all other sampling time points (P < 0.05). The relative abundance of several genera segregated gestation from lactation samples including Roseburia, Prevotella 1, Prevotella 2, Christensenellaceae R-7 group, Ruminococcaceae UCG-002 and Ruminococcaceae UCG-010 (P < 0.01). Furthermore, day 3 was characterised by a significant increase in the relative abundance of Escherichia/Shigella, Fusobacterium and Bacteroides, and a decrease in Alloprevotella, Prevotellaceae UCG-003 and Ruminococcus 1 (P < 0.001). Primiparous sows had overall lower periparturient microbiota diversity (P < 0.01) and there was a significant interaction between parity and sampling time point, with primiparous sows having lower microbiota richness on day -6 (P < 0.001). There was a significant interaction between sow parity and sampling time point on microbiota composition on day -6 and -1 (unweighted UniFrac distances; ≤ 0.01) and day 8 (weighted and unweighted UniFrac distances; P < 0.05). Whilst no significant interactions between sow parity and sampling day were observed for genera relative abundances, multiparous sows had a significantly higher relative abundance of Bacteroidetes dgA-11 gut group and Prevotellaceae UCG-004 (P < 0.01). This study demonstrates that the sow microbiota undergoes longitudinal changes, which are collectively related to periparturient changes in the sow environment, diet and physiological changes to support foetal growth, delivery and the onset of lactation, but also sow parity.
Collapse
Affiliation(s)
- C H Gaukroger
- School of Natural and Environmental Sciences, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK.
| | - S A Edwards
- School of Natural and Environmental Sciences, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - J Walshaw
- Fera Science Limited, York, YO41 1LZ, UK
| | - A Nelson
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, NE1 8ST, UK
| | - I P Adams
- Fera Science Limited, York, YO41 1LZ, UK
| | - C J Stewart
- Clinical and Translational Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - I Kyriazakis
- School of Natural and Environmental Sciences, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| |
Collapse
|
39
|
Soderborg TK, Carpenter CM, Janssen RC, Weir TL, Robertson CE, Ir D, Young BE, Krebs NF, Hernandez TL, Barbour LA, Frank DN, Kroehl M, Friedman JE. Gestational Diabetes Is Uniquely Associated With Altered Early Seeding of the Infant Gut Microbiota. Front Endocrinol (Lausanne) 2020; 11:603021. [PMID: 33329403 PMCID: PMC7729132 DOI: 10.3389/fendo.2020.603021] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/03/2020] [Indexed: 12/17/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is a worldwide public health problem affecting up to 27% of pregnancies with high predictive values for childhood obesity and inflammatory diseases. Compromised seeding of the infant gut microbiota is a risk factor for immunologic and metabolic diseases in the offspring; however, how GDM along with maternal obesity interact to alter colonization remains unknown. We hypothesized that GDM individually and in combination with maternal overweight/obesity would alter gut microbial composition, diversity, and short-chain fatty acid (SCFA) levels in neonates. We investigated 46 full-term neonates born to normal-weight or overweight/obese mothers with and without GDM, accounting for confounders including cesarean delivery, lack of breastfeeding, and exposure to antibiotics. Gut microbiota in 2-week-old neonates born to mothers with GDM exhibited differences in abundance of 26 microbial taxa; 14 of which showed persistent differential abundance after adjusting for pre-pregnancy BMI. Key pioneering gut taxa, including potentially important taxa for establishing neonatal immunity, were reduced. Lactobacillus, Flavonifractor, Erysipelotrichaceae, and unspecified families in Gammaproteobacteria were significantly reduced in neonates from mothers with GDM. GDM was associated with an increase in microbes involved in suppressing early immune cell function (Phascolarctobacterium). No differences in infant stool SCFA levels by maternal phenotype were noted; however, significant correlations were found between microbial abundances and SCFA levels in neonates. Our results suggest that GDM alone and together with maternal overweight/obesity uniquely influences seeding of specific infant microbiota in patterns that set the stage for future risk of inflammatory and metabolic disease.
Collapse
Affiliation(s)
- Taylor K. Soderborg
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Charles M. Carpenter
- Division of Biostatistics and Epidemiology, University of Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Rachel C. Janssen
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Tiffany L. Weir
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, United States
| | - Charles E. Robertson
- Department of Medicine, Division of Infectious Disease, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Diana Ir
- Department of Medicine, Division of Infectious Disease, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Bridget E. Young
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Nancy F. Krebs
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Teri L. Hernandez
- Department of Medicine, Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- College of Nursing, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Linda A. Barbour
- Department of Medicine, Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Daniel N. Frank
- Department of Medicine, Division of Infectious Disease, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Miranda Kroehl
- Division of Biostatistics and Epidemiology, University of Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jacob E. Friedman
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Medicine, Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
40
|
Khine WWT, Rahayu ES, See TY, Kuah S, Salminen S, Nakayama J, Lee YK. Indonesian children fecal microbiome from birth until weaning was different from microbiomes of their mothers. Gut Microbes 2020; 12:1761240. [PMID: 32453977 PMCID: PMC7524161 DOI: 10.1080/19490976.2020.1761240] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Gastrointestinal (GI) microbiota play an important role in human health and wellbeing and the first wave of gut microbes arrives mostly through vertical transmission from mother to child. This study has undertaken to understand the microbiota profile of healthy Southeast Asian mother-infant pairs. Here, we examined the fecal, vaginal and breast milk microbiota of Indonesian mothers and the fecal microbiota of their children from less than 1 month to 48 months old. To determine the immune status of children and the effect of diet at different ages, we examined the level of cytokines, bile acids in the fecal water and weaning food frequency. The fecal microbiota of the children before weaning contained mainly Bacteroides and Bifidobacterium, which presented at low abundance in the samples of mothers. After weaning, the fecal microbiome of children was mainly of the Prevotella type, with decreasing levels of Bifidobacterium, thus becoming more like the fecal microbiome of the mother. The abundance of infant fecal commensals generally correlated inversely with potential pathogens before weaning. The fecal Bifidobacterium in children correlated inversely with the consumption of complex carbohydrates and fruits after weaning. The specific cytokines related to the proliferation and maturation of immunity were found to increase after weaning. A decreasing level of primary bile acids and an increase of secondary bile acids were observed after weaning. This study highlights the change in the GI microbiota of infants to adult-type microbiota after weaning and identifies diet as a major contributing factor.
Collapse
Affiliation(s)
- Wei Wei Thwe Khine
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,Functional Foods Forum, Faculty of Medicine, University of Turku, Turku, Finland
| | - Endang Sutriswati Rahayu
- Faculty of Agricultural Technology and Center for Food & Nutrition Studies, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Ting Yi See
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sherwin Kuah
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Seppo Salminen
- Functional Foods Forum, Faculty of Medicine, University of Turku, Turku, Finland
| | - Jiro Nakayama
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Yuan-Kun Lee
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,Department of Surgery, National University Hospital, Singapore,CONTACT Yuan-Kun Lee Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2117545, Singapore
| |
Collapse
|
41
|
Abbasi A, Aghebati-Maleki A, Yousefi M, Aghebati-Maleki L. Probiotic intervention as a potential therapeutic for managing gestational disorders and improving pregnancy outcomes. J Reprod Immunol 2020; 143:103244. [PMID: 33186834 DOI: 10.1016/j.jri.2020.103244] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/25/2020] [Indexed: 02/06/2023]
Abstract
Recent molecular investigations have significantly developed our knowledge of the characteristics of the reproductive microbiome and their associations with host responses to provide an ideal milieu for the development of the embryo during the peri-implantation period and throughout pregnancy as well as to provide a successful in vitro fertilization and appropriate reproductive outcomes. In this context, the establishment of microbial homeostasis in the female reproductive tract, in various physiological periods, is a substantial challenge, which appears the application of probiotics can facilitate the achievement of this goal. So that, currently, probiotics due to its safe and natural features can be considered as a novel biotherapeutic approach. In this review, we comprehensively discuss the bacterial, fungal, and viral diversity detected in the reproductive tract, and their associations with the establishment of dysbiosis/eubiosis conditions as well as we present the significant outcomes on probiotic intervention as an efficient biotherapeutic strategy for management of gestational disorders and improve pregnancy outcomes.
Collapse
Affiliation(s)
- Amin Abbasi
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz, Iran; Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Aghebati-Maleki
- Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Science, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
42
|
Pirr S, Viemann D. Host Factors of Favorable Intestinal Microbial Colonization. Front Immunol 2020; 11:584288. [PMID: 33117398 PMCID: PMC7576995 DOI: 10.3389/fimmu.2020.584288] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/09/2020] [Indexed: 12/15/2022] Open
Abstract
Gut microbial colonization starts with birth and initiates a complex process between the host and the microbiota. Successful co-development of both establishes a symbiotic mutual relationship and functional homeostasis, while alterations thereof predispose the individual life-long to inflammatory and metabolic diseases. Multiple data have been provided how colonizing microbes induce a reprogramming and maturation of immunity by providing crucial instructing information to the newborn immune system. Less is known about what host factors have influence on the interplay between intestinal immunity and the composition of the gut microbial ecology. Here we review existing evidence regarding host factors that contribute to a favorable development of the gut microbiome and thereby successful maturation of gut mucosal immunity.
Collapse
Affiliation(s)
- Sabine Pirr
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hanover, Germany.,PRIMAL Consortium, Hanover, Germany
| | - Dorothee Viemann
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hanover, Germany.,PRIMAL Consortium, Hanover, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hanover, Germany
| |
Collapse
|
43
|
Offspring susceptibility to metabolic alterations due to maternal high-fat diet and the impact of inhaled ozone used as a stressor. Sci Rep 2020; 10:16353. [PMID: 33004997 PMCID: PMC7530537 DOI: 10.1038/s41598-020-73361-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/08/2020] [Indexed: 12/30/2022] Open
Abstract
The influence of maternal high-fat diet (HFD) on metabolic response to ozone was examined in Long-Evans rat offspring. F0 females were fed control diet (CD; 10%kcal from fat) or HFD (60%kcal from fat) starting at post-natal day (PND) 30. Rats were bred on PND 72. Dietary regimen was maintained until PND 30 when all offspring were switched to CD. On PND 40, F1 offspring (n = 10/group/sex) were exposed to air or 0.8 ppm ozone for 5 h. Serum samples were collected for global metabolomic analysis (n = 8/group/sex). Offspring from HFD dams had increased body fat and weight relative to CD. Metabolomic analysis revealed significant sex-, diet-, and exposure-related changes. Maternal HFD increased free fatty acids and decreased phospholipids (male > female) in air-exposed rats. Microbiome-associated histidine and tyrosine metabolites were increased in both sexes, while 1,5-anhydroglucitol levels decreased in males indicating susceptibility to insulin resistance. Ozone decreased monohydroxy fatty acids and acyl carnitines and increased pyruvate along with TCA cycle intermediates in females (HFD > CD). Ozone increased various amino acids, polyamines, and metabolites of gut microbiota in HFD female offspring indicating gut microbiome alterations. Collectively, these data suggest that maternal HFD increases offspring susceptibility to metabolic alterations in a sex-specific manner when challenged with environmental stressors.
Collapse
|
44
|
Hopson LM, Singleton SS, David JA, Basuchoudhary A, Prast-Nielsen S, Klein P, Sen S, Mazumder R. Bioinformatics and machine learning in gastrointestinal microbiome research and clinical application. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 176:141-178. [PMID: 33814114 DOI: 10.1016/bs.pmbts.2020.08.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The scientific community currently defines the human microbiome as all the bacteria, viruses, fungi, archaea, and eukaryotes that occupy the human body. When considering the variable locations, composition, diversity, and abundance of our microbial symbionts, the sheer volume of microorganisms reaches hundreds of trillions. With the onset of next generation sequencing (NGS), also known as high-throughput sequencing (HTS) technologies, the barriers to studying the human microbiome lowered significantly, making in-depth microbiome research accessible. Certain locations on the human body, such as the gastrointestinal, oral, nasal, and skin microbiomes have been heavily studied through community-focused projects like the Human Microbiome Project (HMP). In particular, the gastrointestinal microbiome (GM) has received significant attention due to links to neurological, immunological, and metabolic diseases, as well as cancer. Though HTS technologies allow deeper exploration of the GM, data informing the functional characteristics of microbiota and resulting effects on human function or disease are still sparse. This void is compounded by microbiome variability observed among humans through factors like genetics, environment, diet, metabolic activity, and even exercise; making GM research inherently difficult to study. This chapter describes an interdisciplinary approach to GM research with the goal of mitigating the hindrances of translating findings into a clinical setting. By applying tools and knowledge from microbiology, metagenomics, bioinformatics, machine learning, predictive modeling, and clinical study data from children with treatment-resistant epilepsy, we describe a proof-of-concept approach to clinical translation and precision application of GM research.
Collapse
Affiliation(s)
- Lindsay M Hopson
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, DC, United States; The McCormick Genomic and Proteomic Center, The George Washington University, Washington, DC, United States; The McCormick Genomic and Proteomic Center, The George Washington University, Washington, DC, United States
| | - Stephanie S Singleton
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, DC, United States
| | - John A David
- Department of Applied Mathematics, Virginia Military Institute, Lexington, VA, United States
| | - Atin Basuchoudhary
- Department of Economics and Business, Virginia Military Institute, Lexington, VA, United States
| | - Stefanie Prast-Nielsen
- Center for Translational Microbiome Research (CTMR), Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, United States
| | - Sabyasachi Sen
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, DC, United States; Department of Medicine, The George Washington University, Washington, DC, United States
| | - Raja Mazumder
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, DC, United States; The McCormick Genomic and Proteomic Center, The George Washington University, Washington, DC, United States.
| |
Collapse
|
45
|
Nyangahu DD, Darby M, Havyarimana E, Brown BP, Horsnell W, Jaspan HB. Preconception helminth infection alters offspring microbiota and immune subsets in a mouse model. Parasite Immunol 2020; 42:e12721. [PMID: 32277499 PMCID: PMC7423732 DOI: 10.1111/pim.12721] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/12/2020] [Accepted: 03/31/2020] [Indexed: 12/17/2022]
Abstract
Both maternal microbiota and helminth infection may alter offspring immunity but the relationship between these is underexplored. We hypothesized that maternal helminth exposure prior to pregnancy has lasting consequences on offspring intestinal microbiota and consequent immunity. Female BALB/c adult mice were infected with 500L3 Nippostrongylus brasiliensis (N brasiliensis). Infection was cleared by ivermectin treatment, and mice were mated 3 weeks post-infection (NbM). Control mice were not infected but were exposed to ivermectin (NvM). We analysed maternal gut microbiota during pregnancy, breastmilk microbiota and offspring faecal microbiota and immunity 2 weeks after delivery. During pregnancy, NbM (Mothers previously infected with Nippostrongylus brasiliensis) displayed significantly altered stool bacterial communities (R2 = .242; P = .001), with increased abundance of Enterococcaceae versus NvM (Naive mothers). Similarly, we observed a profound impact on breastmilk microbiota in NbM vs NvM. Moreover, NbM pups showed significantly altered gut microbial communities at 14 days of age versus those born to NvM with increased relative abundance of Coriobacteriaceae and Micrococcaceae. These changes were associated with alterations in pup immunity including increased frequencies and numbers of activated CD4 T cells (CD4 + CD44hi) in NbM offspring spleens. Taken together, we show that preconception helminth infections impact offspring immunity possibly through alteration of maternal and offspring microbiota.
Collapse
Affiliation(s)
- Donald D Nyangahu
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, RSA
- Departments of Pediatrics and Global Health, University of Washington, Seattle, WA
| | - Matthew Darby
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, RSA
| | - Enock Havyarimana
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, RSA
| | - Bryan P Brown
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle WA, USA
| | - William Horsnell
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, RSA
- Institute of Microbiology and Infection, University of Birmingham, B15 2TT Birmingham, UK
| | - Heather B. Jaspan
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, RSA
- Departments of Pediatrics and Global Health, University of Washington, Seattle, WA
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle WA, USA
| |
Collapse
|
46
|
|
47
|
Camacho JH, Rusinky Pinilla L, Salazar Peña D, Sanabria Dueñas S, Rojas Carvajal D, Burbano Castillo N, Ruiz Peña R, Palacino Saenz I, Martínez Quesada S, García Salazar A, Abdala Galvis N. Microbiota intestinal en pediatría. REPERTORIO DE MEDICINA Y CIRUGÍA 2020. [DOI: 10.31260/repertmedcir.01217372.1100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
La microbiota intestinal es el conjunto de millones de microrganismos vivos ubicados en el tracto gastrointestinal. Es indispensable en múltiples funciones del organismo, regulación de la inmunidad, en aspectos nutricionales y procesos de inflamación sistémica entre otros. La disbiosis es la alteración del equilibrio de la microbiota normal, debido a cambios en la composición, funcionamiento, orden o su distribución; esto puede predisponer al individuo a la adquisición de enfermedades gastrointestinales, alérgicas y metabólicas, entre otras. El objetivo del presente artículo es realizar una revisión narrativa de la literatura sobre los conceptos claves de la microbiota intestinal, sus asociaciones fisiopatológicas con desórdenes gastrointestinales, alérgicos y metabólicos en pediatría.
Collapse
|
48
|
The Infant Gut Microbiota and Risk of Asthma: The Effect of Maternal Nutrition during Pregnancy and Lactation. Microorganisms 2020; 8:microorganisms8081119. [PMID: 32722458 PMCID: PMC7466123 DOI: 10.3390/microorganisms8081119] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
Research has amply demonstrated that early life dysbiosis of the gut microbiota influences the propensity to develop asthma. The influence of maternal nutrition on infant gut microbiota is therefore of growing interest. However, a handful of prospective studies have examined the role of maternal dietary patterns during pregnancy in influencing the infant gut microbiota but did not assess whether this resulted in an increased risk of asthma later in life. The mechanisms involved in the process are also, thus far, poorly documented. There have also been few studies examining the effect of maternal dietary nutrient intake during lactation on the milk microbiota, the effect on the infant gut microbiota and, furthermore, the consequences for asthma development remain largely unknown. Therefore, the specific aim of this mini review is summarizing the current knowledge regarding the effect of maternal nutrition during pregnancy and lactation on the infant gut microbiota composition, and whether it has implications for asthma development.
Collapse
|
49
|
Dai X, Guo Z, Chen D, Li L, Song X, Liu T, Jin G, Li Y, Liu Y, Ajiguli A, Yang C, Wang B, Cao H. Maternal sucralose intake alters gut microbiota of offspring and exacerbates hepatic steatosis in adulthood. Gut Microbes 2020; 11:1043-1063. [PMID: 32228300 PMCID: PMC7524393 DOI: 10.1080/19490976.2020.1738187] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is considered to be associated with diet and gut dysbiosis. Excessive sucralose can induce gut dysbiosis and negatively affect host health. Maternal diet shapes the microbial communities of neonate and this effect continues in later life. We aimed to investigate the effects of maternal sucralose (MS) intake on the susceptibility of offspring to hepatic steatosis in adulthood. METHODS C57BL/6 pregnant mice were randomized into MS group (MS during gestation and lactation) and maternal control (MC) group (MC diet). After weaning, all offspring were fed a control diet until 8 weeks of age, and then treated with a high-fat diet (HFD) for 4 weeks. The intestinal development, mucosal barrier function, and gut microbiota were assessed in the 3-week-old offspring. Moreover, the severity of hepatic steatosis, serum biochemistry, lipid metabolism, and gut microbiota was then assessed in the 12th week. RESULTS MS significantly inhibited intestinal development and disrupted barrier function in 3-week-old offspring. MS also induced intestinal low-grade inflammation, significantly changed the compositions and diversity of gut microbiota including reducing butyrate-producing bacteria and cecal butyrate production with down-regulation of GPR43. Mechanically, blocking GPR43 blunted the anti-inflammatory effect of one of the butyrate-producing bacteria, Clostridium butyricum in vitro. After HFD treatment, MS exacerbated hepatic steatosis, and disturbed fatty acid biosynthesis and metabolism, accompanied by inducing gut dysbiosis compared with MC group. CONCLUSIONS MS intake inhibits intestinal development, induces gut dysbiosis in offspring through down-regulation of GPR43, and exacerbates HFD-induced hepatic steatosis in adulthood.
Collapse
Affiliation(s)
- Xin Dai
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Zixuan Guo
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Danfeng Chen
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Lu Li
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Xueli Song
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Ge Jin
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Yun Li
- Department of Pharmacy, General Hospital, Tianjin Medical University, Tianjin, China
| | - Yi Liu
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin, China,Department of Gastroenterology and Hepatology, Hotan District People’s Hospital, Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Aihemaiti Ajiguli
- Department of Gastroenterology and Hepatology, Hotan District People’s Hospital, Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Cheng Yang
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine,Tianjin
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China,CONTACT Bangmao Wang Department of Gastroenterology and HepatologyGeneral Hospital, Tianjin Medical University, 154 Anshan Road, Heping District, China, Tianjin , 300052
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China,Department of Gastroenterology and Hepatology, Hotan District People’s Hospital, Xinjiang Uygur Autonomous Region, Xinjiang, China,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine,Tianjin,Hailong Cao Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, 154 Anshan Road, Heping District, Tianjin300052, China
| |
Collapse
|
50
|
Abstract
The causes of neonatal gut injury are multifactorial and include ischemia, tissue hypoxia due to anemia, excessive inflammation, deficiency of growth factors, and food protein sensitivity. The developing intestinal microbiome plays a role in some of these forms of intestinal injury but knowledge of its relative role in each remains poorly understood. Commensal bacteria are required for normal immune development and immune tolerance. Dysbiosis in the neonatal gut that alters the patterns of commensal and pathogenic bacteria may accentuate gut injury.
Collapse
Affiliation(s)
- Mohan Pammi
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, 6621, Fannin, WT 6-104, Houston, TX 77030 USA.
| | - Emily Hollister
- Diversigen, Inc, Information Technology and Analytics, 2450 Holcombe Boulevard, Suite BCMA, Houston, TX 77021, USA
| | - Josef Neu
- Section of Neonatology, Department of Pediatrics, University of Florida, 1600 SW Archer Road, Gainesville, FL 32610, USA
| |
Collapse
|