1
|
Momeni N, Mousavi SN, Chiti H, Heidarzadeh S. A maternal sweet diet is associated with the gut dysbiosis in the first trimester of pregnancy. BMC Nutr 2024; 10:162. [PMID: 39695908 DOI: 10.1186/s40795-024-00972-5] [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: 08/17/2024] [Accepted: 12/08/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The composition of maternal gut phylum in each trimester of pregnancy has been associated with fetal development, separately. Diet is a main effective factor on the gut composition of phylum. However, associations between dietary glycemic index (GI), load (GL) and total antioxidant capacity (TAC) not studied with the gut population of phylum in mothers at the first trimester of pregnancy. MATERIALS AND METHODS Ninety healthy pregnant women aged 18-40 yrs, in the first trimester, were participated. Stool samples were gathered in a fasting state. Population of dominant phylum was determined after DNA extraction based on the 16SrRNA expression, as a housekeeping gene. Dietary intake was collected by a validated food frequency questionnaire and dietary indices were computed. RESULTS The Proteobacteria population was significantly higher in the gut of pregnant mothers than the other phylum (p < 0.001). Participants in the highest level of dietary GI had lower Bacteroidetes (p < 0.001) and Actinobacteria (p = 0.04) in their gut compared to the lowest level. Participants in the lowest level of dietary GL had higher Bacteroidetes (p < 0.001) and lower proteobacteria (p = 0.04) in their gut than the highest level. Dietary selenium showed a significant negative effect on the Firmicutes (p = 0.04) and Proteobacteria (p = 0.04), however positively affected the Actinobacteria (p = 0.01) population. Dietary zinc and manganese showed a negative effect on the Firmicutes population (p = 0.01 and p = 0.003). Zinc and vitamin E showed a negative effect on the Proteobacteria population (p = 0.04 and p = 0.03). CONCLUSIONS A maternal diet with high GI and GL have been associated with the gut dysbiosis, however dietary intake of selenium, zinc, manganese and vitamin E act in favor of the intestinal eubiosis in the first trimester of pregnancy.
Collapse
Affiliation(s)
- Navid Momeni
- Zanjan Metabolic Diseases Research Center, Health and Metabolic Diseases Research Institute, Zanjan University of Medical Sciences, Parvin Etesami St, Azadi Square, Zanjan, Iran
| | - Seyedeh Neda Mousavi
- Zanjan Metabolic Diseases Research Center, Health and Metabolic Diseases Research Institute, Zanjan University of Medical Sciences, Parvin Etesami St, Azadi Square, Zanjan, Iran.
- Department of Nutrition, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Hossein Chiti
- Zanjan Metabolic Diseases Research Center, Health and Metabolic Diseases Research Institute, Zanjan University of Medical Sciences, Parvin Etesami St, Azadi Square, Zanjan, Iran.
| | - Siamak Heidarzadeh
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
2
|
Sun H, Chen M, Liao J, He L, Wan B, Yin J, Zhang X. The maternal lifestyle in pregnancy: Implications for foetal skeletal muscle development. J Cachexia Sarcopenia Muscle 2024; 15:1641-1650. [PMID: 39155495 PMCID: PMC11446712 DOI: 10.1002/jcsm.13556] [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: 04/29/2024] [Revised: 06/20/2024] [Accepted: 07/15/2024] [Indexed: 08/20/2024] Open
Abstract
The world is facing a global nutrition crisis, as evidenced by the rising incidence of metabolic disorders such as obesity, insulin resistance and chronic inflammation. Skeletal muscle is the largest tissue in humans and plays an important role in movement and host metabolism. Muscle fibre formation occurs mainly during the embryonic stage. Therefore, maternal lifestyle, especially nutrition and exercise during pregnancy, has a critical influence on foetal skeletal muscle development and the subsequent metabolic health of the offspring. In this review, the influence of maternal obesity, malnutrition and micronutrient intake on foetal skeletal muscle development is systematically summarized. We also aim to describe how maternal exercise shapes foetal muscle development and metabolic health in the offspring. The role of maternal gut microbiota and its metabolites on foetal muscle development is further discussed, although this field is still in its 'infancy'. This review will provide new insights to reduce the global crisis of metabolic disorders and highlight current gaps to promote further research.
Collapse
Affiliation(s)
- Haijun Sun
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Meixia Chen
- Institute of Animal Husbandry and Veterinary MedicineBeijing Academy of Agriculture and Forestry SciencesBeijingChina
| | - Jialong Liao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Linjuan He
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Boyang Wan
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Jingdong Yin
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
- Frontiers Science Center for Molecular Design Breeding (MOE)BeijingChina
| | - Xin Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
- Frontiers Science Center for Molecular Design Breeding (MOE)BeijingChina
| |
Collapse
|
3
|
Baptista FI, Ambrósio AF. Tracing the influence of prenatal risk factors on the offspring retina: Focus on development and putative long-term consequences. Eur J Clin Invest 2024; 54:e14266. [PMID: 38864773 DOI: 10.1111/eci.14266] [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: 03/12/2024] [Revised: 05/10/2024] [Accepted: 05/29/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Pregnancy represents a window of vulnerability to fetal development. Disruptions in the prenatal environment during this crucial period can increase the risk of the offspring developing diseases over the course of their lifetime. The central nervous system (CNS) has been shown to be particularly susceptible to changes during crucial developmental windows. To date, research focused on disruptions in the development of the CNS has predominantly centred on the brain, revealing a correlation between exposure to prenatal risk factors and the onset of neuropsychiatric disorders. Nevertheless, some studies indicate that the retina, which is part of the CNS, is also vulnerable to in utero alterations during pregnancy. Such changes may affect neuronal, glial and vascular components of the retina, compromising retinal structure and function and possibly impairing visual function. METHODS A search in the PubMed database was performed, and any literature concerning prenatal risk factors (drugs, diabetes, unbalanced diet, infection, glucocorticoids) affecting the offspring retina were included. RESULTS This review collects evidence on the cellular, structural and functional changes occurring in the retina triggered by maternal risk factors during pregnancy. We highlight the adverse impact on retinal development and its long-lasting effects, providing a critical analysis of the current knowledge while underlining areas for future research. CONCLUSIONS Appropriate recognition of the prenatal risk factors that negatively impact the developing retina may provide critical clues for the design of preventive strategies and for early therapeutic intervention that could change retinal pathology in the progeny.
Collapse
Affiliation(s)
- Filipa I Baptista
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - António F Ambrósio
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image, Coimbra, Portugal
| |
Collapse
|
4
|
Sha Y, Liu X, Li X, Wang Z, Shao P, Jiao T, He Y, Zhao S. Succession of rumen microbiota and metabolites across different reproductive periods in different sheep breeds and their impact on the growth and development of offspring lambs. MICROBIOME 2024; 12:172. [PMID: 39267132 PMCID: PMC11397069 DOI: 10.1186/s40168-024-01892-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 07/30/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND The microbiota and metabolites in the gastrointestinal tracts of female animals at different reproductive periods are very important to the growth, development, and health of themselves and their offspring. However, the changes in the gastrointestinal microbiota and metabolites throughout reproductive period of different sheep breeds and their effects on the growth and development of offspring lambs are still unclear. Hence, this study presents an assessment of the reproductive hormone levels, immune levels, rumen microbiota, and metabolites in Hu sheep and Suffolk ewes at different reproductive periods and their effects on the growth and development of offspring lambs. RESULTS Hu sheep and Suffolk during non-pregnancy, pregnancy, and lactation were used as the research objects to determine reproductive and immune indexes of ewes at different periods, analyze rumen microbiome and metabolome, and track the growth performance and development of offspring lambs. The results showed that the reproductive hormone and immune levels of Hu sheep and Suffolk underwent adaptive changes across different reproductive periods. Compared with non-pregnancy, the microbial energy metabolism and lipid metabolism function decreased during Hu sheep pregnancy, and energy metabolism function decreased during lactation. In Suffolk, energy metabolism, glycan biosynthesis, and metabolism function were enhanced during pregnancy, and the metabolism of cofactors and vitamins was enhanced during lactation. Prevotella increased in Suffolk during pregnancy and lactation (P < 0.05) and was positively correlated with the birth weight and body size of the lambs (P < 0.05). Moreover, the abundances of Butyrivibrio and Rikenellaceae_RC9_gut_group during pregnancy were positively correlated with the intestinal immunity of the offspring lambs (P < 0.05), thereby regulating the intestinal immunity level of the lambs. Metabolomic analysis revealed that the protein digestion, absorption, and amino acid metabolism of Hu sheep were enhanced during pregnancy, which provided amino acids for the growth and development of pregnant ewes and fetuses and was significantly correlated with the birth weight, body size, and intestinal immunity of lambs (P < 0.05). Simultaneously, there was an increase in acetate and propionate during the pregnancy and lactation period of both Hu sheep and Suffolk, providing energy for ewes during reproductive period. Moreover, the microbiota during the lactation period was significantly correlated with the milk quality and lambs daily gain (P < 0.05). CONCLUSIONS This study revealed the characteristic succession changes in the rumen microbiota and its metabolites at different reproductive periods in sheep breeds and their regulation of reproductive hormone and immune levels and identified their potential effects on the growth and development of offspring lambs. The findings provide valuable insights into the health and feeding management of different sheep breeds during the reproductive stage. Video Abstract.
Collapse
Affiliation(s)
- Yuzhu Sha
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Xiu Liu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Xiongxiong Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Zhengwen Wang
- College of Pratacultural Science, Gansu Agricultural University / Key Laboratory for Grassland Ecosystem, Ministry of Education / Sino-US Grassland Animal Husbandry Sustainable Development Research Center, Lanzhou, 730070, China
| | - Pengyang Shao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Ting Jiao
- College of Pratacultural Science, Gansu Agricultural University / Key Laboratory for Grassland Ecosystem, Ministry of Education / Sino-US Grassland Animal Husbandry Sustainable Development Research Center, Lanzhou, 730070, China
| | - Yanyu He
- School of Fundamental Sciences, Massey University, Palmerston North, 4410, New Zealand
| | - Shengguo Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China.
| |
Collapse
|
5
|
Bonet ML, Ribot J, Sánchez J, Palou A, Picó C. Early Life Programming of Adipose Tissue Remodeling and Browning Capacity by Micronutrients and Bioactive Compounds as a Potential Anti-Obesity Strategy. Cells 2024; 13:870. [PMID: 38786092 PMCID: PMC11120104 DOI: 10.3390/cells13100870] [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: 04/09/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
The early stages of life, especially the period from conception to two years, are crucial for shaping metabolic health and the risk of obesity in adulthood. Adipose tissue (AT) plays a crucial role in regulating energy homeostasis and metabolism, and brown AT (BAT) and the browning of white AT (WAT) are promising targets for combating weight gain. Nutritional factors during prenatal and early postnatal stages can influence the development of AT, affecting the likelihood of obesity later on. This narrative review focuses on the nutritional programming of AT features. Research conducted across various animal models with diverse interventions has provided insights into the effects of specific compounds on AT development and function, influencing the development of crucial structures and neuroendocrine circuits responsible for energy balance. The hormone leptin has been identified as an essential nutrient during lactation for healthy metabolic programming against obesity development in adults. Studies have also highlighted that maternal supplementation with polyunsaturated fatty acids (PUFAs), vitamin A, nicotinamide riboside, and polyphenols during pregnancy and lactation, as well as offspring supplementation with myo-inositol, vitamin A, nicotinamide riboside, and resveratrol during the suckling period, can impact AT features and long-term health outcomes and help understand predisposition to obesity later in life.
Collapse
Affiliation(s)
- M. Luisa Bonet
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (M.L.B.); (J.S.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Artificial Intelligence Research Institute of the Balearic Islands (IAIB), University of the Balearic Islands, 07122 Palma, Spain
| | - Joan Ribot
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (M.L.B.); (J.S.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Juana Sánchez
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (M.L.B.); (J.S.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (M.L.B.); (J.S.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Artificial Intelligence Research Institute of the Balearic Islands (IAIB), University of the Balearic Islands, 07122 Palma, Spain
| | - Catalina Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (M.L.B.); (J.S.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Artificial Intelligence Research Institute of the Balearic Islands (IAIB), University of the Balearic Islands, 07122 Palma, Spain
| |
Collapse
|
6
|
Guo W, Liu S, Khan MZ, Wang J, Chen T, Alugongo GM, Li S, Cao Z. Bovine milk microbiota: Key players, origins, and potential contributions to early-life gut development. J Adv Res 2024; 59:49-64. [PMID: 37423549 PMCID: PMC11081965 DOI: 10.1016/j.jare.2023.06.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/11/2023] Open
Abstract
BACKGROUND Bovine milk is a significant substitute for human breast milk and holds great importance in infant nutrition and health. Apart from essential nutrients, bovine milk also contains bioactive compounds, including a microbiota derived from milk itself rather than external sources of contamination. AIM OF REVIEW Recognizing the profound impact of bovine milk microorganisms on future generations, our review focuses on exploring their composition, origins, functions, and applications. KEY SCIENTIFIC CONCEPTS OF REVIEW Some of the primary microorganisms found in bovine milk are also present in human milk. These microorganisms are likely transferred to the mammary gland through two pathways: the entero-mammary pathway and the rumen-mammary pathway. We also elucidated potential mechanisms by which milk microbiota contribute to infant intestinal development. The mechanisms include the enhancing of the intestinal microecological niche, promoting the maturation of immune system, strengthening the intestinal epithelial barrier function, and interacting with milk components (e.g., oligosaccharides) via cross-feeding effect. However, given the limited understanding of bovine milk microbiota, further studies are necessary to validate hypotheses regarding their origins and to explore their functions and potential applications in early intestinal development.
Collapse
Affiliation(s)
- Wenli Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shuai Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Muhammad Z Khan
- Faculty of Veterinary and Animal Sciences, Department of Animal Breeding and Genetics, The University of Agriculture, Dera Ismail Khan 29220, Pakistan
| | - Jingjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Tianyu Chen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Gibson M Alugongo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shengli Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zhijun Cao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
7
|
Adamczak AM, Werblińska A, Jamka M, Walkowiak J. Maternal-Foetal/Infant Interactions-Gut Microbiota and Immune Health. Biomedicines 2024; 12:490. [PMID: 38540103 PMCID: PMC10967760 DOI: 10.3390/biomedicines12030490] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 01/03/2025] Open
Abstract
In recent years, the number of scientific publications on the role of intestinal microbiota in shaping human health, as well as the occurrence of intestinal dysbiosis in various disease entities, has increased dynamically. However, there is a gap in comprehensively understanding the factors influencing a child's gut microbiota. This review discusses the establishment of gut microbiota and the immunological mechanisms regulating children's microbiota, emphasising the importance of prioritising the development of appropriate gut microbiota in a child from the planning stages of pregnancy. The databases PubMed, Web of Sciences, Cochrane, Scopus and Google Scholar were searched to identify relevant articles. A child's gut microbiota composition is influenced by numerous factors, such as diet during pregnancy, antibiotic therapy, the mother's vaginal microbiota, delivery method, and, later, feeding method and environmental factors. During pregnancy, the foetus naturally acquires bacterial strains from the mother through the placenta, thereby shaping the newborn's immune system. Inappropriate maternal vaginal microbiota may increase the risk of preterm birth. Formula-fed infants typically exhibit a more diverse microbiota than their breastfed counterparts. These factors, among others, shape the maturation of the child's immune system, impacting the production of IgA antibodies that are central to cellular humoral immune defence. Further research should focus on identifying specific microbiota-immune system interactions influencing a child's immune health and developing personalised treatment strategies for immune-related disorders.
Collapse
Affiliation(s)
- Ada Maria Adamczak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 27/33 Szpitalna Street, 60-572 Poznań, Poland; (A.M.A.); (M.J.)
| | - Alicja Werblińska
- Greater Poland Centre for Pulmonology and Thoracic Surgery Named after Eugenia and Janusz Zeyland, 62 Szamarzewskiego Street, 60-569 Poznań, Poland;
| | - Małgorzata Jamka
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 27/33 Szpitalna Street, 60-572 Poznań, Poland; (A.M.A.); (M.J.)
| | - Jarosław Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 27/33 Szpitalna Street, 60-572 Poznań, Poland; (A.M.A.); (M.J.)
| |
Collapse
|
8
|
Liang Y, Zhao C, Zhao L, Sheng D, Chen B, Zhao G, Wang Q, Zhang L. Taxonomic and functional shifts of gut microbiome in immunoglobulin A vasculitis children and their mothers. Front Pediatr 2024; 12:1356529. [PMID: 38410769 PMCID: PMC10895042 DOI: 10.3389/fped.2024.1356529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/25/2024] [Indexed: 02/28/2024] Open
Abstract
Objectives To examine the gut microbiota characteristics in children with immunoglobulin A vasculitis and their interrelationships with the host, while evaluate the vertical inheritance of microbiota in the development and progression of IgA vasculitis. Methods This study investigated the gut microbiome of 127 IgA vasculitis mother-child pairs and 62 matched healthy mother-child pairs, and compared the gut microbial composition of different groups. The pathway enrichment analysis evaluated potential gut microbiome-mediated pathways involved in the pathophysiology of IgA vasculitis. The Spearman correlation analysis illustrated the relationships between clinical variables and bacterial biomarkers. Results This study identified distinct intestinal microbiome in IgA vasculitis children compared to healthy children, and further pointed out the association in gut microbiota between IgA vasculitis children's and their mother's. The relative abundance of Megamonas and Lactobacillus in IgAV children was positively correlated with that in their mothers. The pathway enrichment analysis found microbial biosynthesis of vitamins and essential amino acids was upregulated in children with IgA vasculitis. Correlation analysis showed bacterial biomarkers were correlated with indicators of blood coagulation. Conclusion Children with IgA vasculitis have unique bacterial biomarkers and may affect coagulation function, and their gut microbiome was closely associated with that of their mothers. The observed association in gut microbiota between IgA vasculitis children and their mothers suggested a potential intergenerational influence of the maternal microbiota on the development or progression of IgA vasculitis in children.
Collapse
Affiliation(s)
- Yijia Liang
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Changying Zhao
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lanlan Zhao
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dashuang Sheng
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bin Chen
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guoping Zhao
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
- CAS Key Laboratory of Computational Biology, Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qinghua Wang
- School of Biological Science and Technology, University of Jinan, Jinan, China
| | - Lei Zhang
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| |
Collapse
|
9
|
Samarra A, Flores E, Bernabeu M, Cabrera-Rubio R, Bäuerl C, Selma-Royo M, Collado MC. Shaping Microbiota During the First 1000 Days of Life. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1449:1-28. [PMID: 39060728 DOI: 10.1007/978-3-031-58572-2_1] [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: 07/28/2024]
Abstract
Given that the host-microbe interaction is shaped by the immune system response, it is important to understand the key immune system-microbiota relationship during the period from conception to the first years of life. The present work summarizes the available evidence concerning human reproductive microbiota, and also, the microbial colonization during early life, focusing on the potential impact on infant development and health outcomes. Furthermore, we conclude that some dietary strategies including specific probiotics and other-biotics could become potentially valuable tools to modulate the maternal-neonatal microbiota during this early critical window of opportunity for targeted health outcomes throughout the entire lifespan.
Collapse
Affiliation(s)
- Anna Samarra
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna-Valencia, Spain
| | - Eduard Flores
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna-Valencia, Spain
| | - Manuel Bernabeu
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna-Valencia, Spain
| | - Raul Cabrera-Rubio
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna-Valencia, Spain
| | - Christine Bäuerl
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna-Valencia, Spain
| | - Marta Selma-Royo
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna-Valencia, Spain
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna-Valencia, Spain.
| |
Collapse
|
10
|
Koren O, Konnikova L, Brodin P, Mysorekar IU, Collado MC. The maternal gut microbiome in pregnancy: implications for the developing immune system. Nat Rev Gastroenterol Hepatol 2024; 21:35-45. [PMID: 38097774 DOI: 10.1038/s41575-023-00864-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/20/2023] [Indexed: 01/04/2024]
Abstract
The gut microbiome has important roles in host metabolism and immunity, and microbial dysbiosis affects human physiology and health. Maternal immunity and microbial metabolites during pregnancy, microbial transfer during birth, and transfer of immune factors, microorganisms and metabolites via breastfeeding provide critical sources of early-life microbial and immune training, with important consequences for human health. Only a few studies have directly examined the interactions between the gut microbiome and the immune system during pregnancy, and the subsequent effect on offspring development. In this Review, we aim to describe how the maternal microbiome shapes overall pregnancy-associated maternal, fetal and early neonatal immune systems, focusing on the existing evidence and highlighting current gaps to promote further research.
Collapse
Affiliation(s)
- Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Liza Konnikova
- Department of Paediatrics and Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Petter Brodin
- Department of Immunology and Inflammation, Imperial College London, London, UK
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Indira U Mysorekar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain.
| |
Collapse
|
11
|
Wang B, Geng M, Li M, Wang X, Gan H, Tang Y, Yang Q, Liu Y, Yang X, Wang S, Liu K, Wei Z, Shao S, Zhu P, Cao Y, Tao F. Preconception exposure to environmental antibiotics among childbearing couples in Anhui and health risk assessment: A multicenter population-based representative study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 265:115514. [PMID: 37783111 DOI: 10.1016/j.ecoenv.2023.115514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/27/2023] [Accepted: 09/21/2023] [Indexed: 10/04/2023]
Abstract
Only few studies have assessed the health effects due to preconception exposure to antibiotics among childbearing couples. This study investigated the status of preconception exposure to antibiotics among childbearing couples in Anhui, associated with health risks, and influencing factors. Overall, 1500 childbearing couples were randomly selected from the Reproductive Health of Childbearing Couples - Anhui Cohort (RHCC-AC). The urinary levels of 40 antibiotics and 2 metabolites were determined, and specific gravity (SG) adjusted concentrations of antibiotics were measured to assess health risks. Generalized linear models were used to assess the associations of urinary SG-adjusted concentration of antibiotics with demographic parameters and diet frequency. The total detection rates of all antibiotics were 98.9 % and 99.3 % in wives and husbands, respectively. The detection rates of veterinary antibiotics (VAs) and preferred as VAs (PVAs) were above 90 %. Among eight antibiotics, sulfonamides (95.1 %) and fluoroquinolones (87.6 %) had the highest detection rates in couples. Approximately four-fifths of couples were simultaneously exposed to at least three different antibiotics, and more than half of them were exposed to low concentrations of antibiotics. 8.9 % and 9.2 % of wives and husbands had hazard index value of antibiotics exposure greater than 1. Antibiotic concentrations were associated with residence, sampling season, and diet frequency. In Anhui, nearly 98 % of childbearing couples have environmental exposure to antibiotics, and VAs and PVAs are the primary antibiotics. More than 8 % of couples had health risks due to antibiotic exposure. Several potential determinants of urinary antibiotics deserve more attention in future research.
Collapse
Affiliation(s)
- Baolin Wang
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Menglong Geng
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Mengdie Li
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Xiaorui Wang
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Hong Gan
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Ying Tang
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Qianhui Yang
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Yuwei Liu
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Xinliu Yang
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Sheng Wang
- The Center for Scientific Research of Anhui Medical University, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Kaiyong Liu
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Zhaolian Wei
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Shanshan Shao
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Peng Zhu
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Yunxia Cao
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Fangbiao Tao
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China.
| |
Collapse
|
12
|
Strout N, Pasic L, Hicks C, Chua XY, Tashvighi N, Butler P, Liu Z, El-Assaad F, Holmes E, Susic D, Samaras K, Craig ME, Davis GK, Henry A, Ledger WL, El-Omar EM. The MothersBabies Study, an Australian Prospective Cohort Study Analyzing the Microbiome in the Preconception and Perinatal Period to Determine Risk of Adverse Pregnancy, Postpartum, and Child-Related Health Outcomes: Study Protocol. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:6736. [PMID: 37754596 PMCID: PMC10531411 DOI: 10.3390/ijerph20186736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/03/2023] [Accepted: 09/06/2023] [Indexed: 09/28/2023]
Abstract
The microbiome has emerged as a key determinant of human health and reproduction, with recent evidence suggesting a dysbiotic microbiome is implicated in adverse perinatal health outcomes. The existing research has been limited by the sample collection and timing, cohort design, sample design, and lack of data on the preconception microbiome. This prospective, longitudinal cohort study will recruit 2000 Australian women, in order to fully explore the role of the microbiome in the development of adverse perinatal outcomes. Participants are enrolled for a maximum of 7 years, from 1 year preconception, through to 5 years postpartum. Assessment occurs every three months until pregnancy occurs, then during Trimester 1 (5 + 0-12 + 6 weeks gestation), Trimester 2 (20 + 0-24 + 6 weeks gestation), Trimester 3 (32 + 0-36 + 6 weeks gestation), and postpartum at 1 week, 2 months, 6 months, and then annually from 1 to 5 years. At each assessment, maternal participants self-collect oral, skin, vaginal, urine, and stool samples. Oral, skin, urine, and stool samples will be collected from children. Blood samples will be obtained from maternal participants who can access a study collection center. The measurements taken will include anthropometric, blood pressure, heart rate, and serum hormonal and metabolic parameters. Validated self-report questionnaires will be administered to assess diet, physical activity, mental health, and child developmental milestones. Medications, medical, surgical, obstetric history, the impact of COVID-19, living environments, and pregnancy and child health outcomes will be recorded. Multiomic bioinformatic and statistical analyses will assess the association between participants who developed high-risk and low-risk pregnancies, adverse postnatal conditions, and/or childhood disease, and their microbiome for the different sample types.
Collapse
Affiliation(s)
- Naomi Strout
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
| | - Lana Pasic
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
| | - Chloe Hicks
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
| | - Xin-Yi Chua
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
| | - Niki Tashvighi
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
| | - Phoebe Butler
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
| | - Zhixin Liu
- UNSW Stats Central, Biological Sciences South Building (E26), Level 2 Kensington, UNSW Sydney, Sydney, NSW 2052, Australia
- Healthdirect Australia, Level 4, 477 Pitt Street, Sydney, NSW 2000, Australia
| | - Fatima El-Assaad
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
| | - Elaine Holmes
- The Australian National Phenome Centre, Harry Perkins Institute, Murdoch University, Perth, WA 6150, Australia;
| | - Daniella Susic
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
- Department of Women’s and Children’s Health, St George Hospital, Kogarah, NSW 2217, Australia; (G.K.D.); (A.H.)
- Discipline of Women’s Health, School of Clinical Medicine, UNSW Sydney, Sydney, NSW 2052, Australia; (M.E.C.); (W.L.L.)
| | - Katherine Samaras
- Complex Diseases Program, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia;
- Department of Endocrinology, St Vincent’s Hospital, Darlinghurst, NSW 2010, Australia
- St Vincent’s Clinical Campus, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Maria E. Craig
- Discipline of Women’s Health, School of Clinical Medicine, UNSW Sydney, Sydney, NSW 2052, Australia; (M.E.C.); (W.L.L.)
| | - Gregory K. Davis
- Department of Women’s and Children’s Health, St George Hospital, Kogarah, NSW 2217, Australia; (G.K.D.); (A.H.)
- Discipline of Women’s Health, School of Clinical Medicine, UNSW Sydney, Sydney, NSW 2052, Australia; (M.E.C.); (W.L.L.)
| | - Amanda Henry
- Department of Women’s and Children’s Health, St George Hospital, Kogarah, NSW 2217, Australia; (G.K.D.); (A.H.)
- Discipline of Women’s Health, School of Clinical Medicine, UNSW Sydney, Sydney, NSW 2052, Australia; (M.E.C.); (W.L.L.)
| | - William L. Ledger
- Discipline of Women’s Health, School of Clinical Medicine, UNSW Sydney, Sydney, NSW 2052, Australia; (M.E.C.); (W.L.L.)
| | - Emad M. El-Omar
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW 2052, Australia; (N.S.); (L.P.); (C.H.); (X.-Y.C.); (F.E.-A.); (D.S.)
| |
Collapse
|
13
|
Liu T, Jia F, Differding MK, Zhao N, Doyon M, Bouchard L, Perron P, Guérin R, Massé E, Hivert MF, Mueller NT. Pre-pregnancy body mass index and gut microbiota of mothers and children 5 years postpartum. Int J Obes (Lond) 2023; 47:807-816. [PMID: 37173396 PMCID: PMC10911130 DOI: 10.1038/s41366-023-01322-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 04/19/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND Maternal pre-pregnancy body mass index (BMI) has been linked to altered gut microbiota in women shortly after delivery and in their offspring in the first few years of life. But little is known about how long these differences persist. METHODS We followed 180 mothers and children from pregnancy until 5-year postpartum in the Gen3G cohort (Canada, enrolled 2010-2013). At 5 years postpartum we collected stool samples from mothers and children and estimated the gut microbiota by 16 S rRNA sequencing (V4 region) using Illumina MiSeq, and assigning amplicon sequence variants (ASV). We examined whether overall microbiota composition (as measured by microbiota β diversity) was more similar between mother-child pairs compared to between mothers or between children. We also assessed whether mother-child pair sharing of overall microbiota composition differed by the weight status of mothers before pregnancy and of children at 5-year. Furthermore, in mothers, we examined whether pre-pregnancy BMI, BMI 5-year postpartum, and change in BMI between time points was associated with maternal gut microbiota 5-year postpartum. In children, we further examined associations of maternal pre-pregnancy BMI and child 5-year BMI z-score with child 5-year gut microbiota. RESULTS Mother-child pairs had greater similarity in overall microbiome composition compared to between mothers and between children. In mothers, higher pre-pregnancy BMI and 5-year postpartum BMI were associated with lower microbiota observed ASV richness and Chao 1 index; in children's gut microbiota, higher maternal pre-pregnancy BMI was weakly associated with lower microbiota Shannon index, whereas child's 5-year BMI z-score was associated with higher observed ASV richness. Pre-pregnancy BMI was also linked to differential abundances of several microbial ASVs in the Ruminococcaceae and Lachnospiraceae families, but no specific ASV had overlapping associations with BMI measures in both mothers and children. CONCLUSIONS Pre-pregnancy BMI was associated with gut microbiota diversity and composition of mothers and children 5 years after birth, however, the nature and direction of most associations differed for mothers and children. Future studies are encouraged to confirm our findings and look into potential mechanisms or factors that may drive these associations.
Collapse
Affiliation(s)
- Tiange Liu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Fan Jia
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Moira K Differding
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ni Zhao
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Myriam Doyon
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC, Canada
| | - Luigi Bouchard
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC, Canada
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC, Canada
- Department of Medical Biology, CIUSSS-SLSJ, Saguenay, QC, Canada
| | - Patrice Perron
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC, Canada
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Renée Guérin
- Department of Medical Biology, CIUSSS-SLSJ, Saguenay, QC, Canada
| | - Eric Massé
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Marie-France Hivert
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC, Canada
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA, USA
| | - Noel T Mueller
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
14
|
Cerdó T, Nieto-Ruíz A, García-Santos JA, Rodríguez-Pöhnlein A, García-Ricobaraza M, Suárez A, Bermúdez MG, Campoy C. Current Knowledge About the Impact of Maternal and Infant Nutrition on the Development of the Microbiota-Gut-Brain Axis. Annu Rev Nutr 2023; 43:251-278. [PMID: 37603431 DOI: 10.1146/annurev-nutr-061021-025355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
The prenatal and early postnatal periods are stages during which dynamic changes and the development of the brain and gut microbiota occur, and nutrition is one of the most important modifiable factors that influences this process. Given the bidirectional cross talk between the gut microbiota and the brain through the microbiota-gut-brain axis (MGBA), there is growing interest in evaluating the potential effects of nutritional interventions administered during these critical developmental windows on gut microbiota composition and function and their association with neurodevelopmental outcomes. We review recent preclinical and clinical evidence from animal studies and infant/child populations. Although further research is needed, growing evidence suggests that different functional nutrients affect the establishment and development of the microbiota-gut-brain axis and could have preventive and therapeutic use in the treatment of neuropsychiatric disorders. Therefore, more in-depth knowledge regarding the effect of nutrition on the MGBA during critical developmental windows may enable the prevention of later neurocognitive and behavioral disorders and allow the establishment of individualized nutrition-based programs that can be used from the prenatal to the early and middle stages of life.
Collapse
Affiliation(s)
- Tomás Cerdó
- Maimonides Institute for Research in Biomedicine of Córdoba, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
| | - Ana Nieto-Ruíz
- Department of Paediatrics, Faculty of Medicine, University of Granada, Granada, Spain;
- Instituto de Investigación Biosanitaria (IBS-GRANADA), Granada, Spain
- Instituto de Neurociencias "Doctor Federico Olóriz," Biomedical Research Centre, University of Granada, Granada, Spain
| | - José Antonio García-Santos
- Department of Paediatrics, Faculty of Medicine, University of Granada, Granada, Spain;
- Instituto de Investigación Biosanitaria (IBS-GRANADA), Granada, Spain
- Instituto de Neurociencias "Doctor Federico Olóriz," Biomedical Research Centre, University of Granada, Granada, Spain
| | - Anna Rodríguez-Pöhnlein
- Department of Paediatrics, Faculty of Medicine, University of Granada, Granada, Spain;
- Instituto de Investigación Biosanitaria (IBS-GRANADA), Granada, Spain
- Instituto de Neurociencias "Doctor Federico Olóriz," Biomedical Research Centre, University of Granada, Granada, Spain
| | - María García-Ricobaraza
- Department of Paediatrics, Faculty of Medicine, University of Granada, Granada, Spain;
- Instituto de Investigación Biosanitaria (IBS-GRANADA), Granada, Spain
- Instituto de Neurociencias "Doctor Federico Olóriz," Biomedical Research Centre, University of Granada, Granada, Spain
| | - Antonio Suárez
- Department of Biochemistry and Molecular Biology 2, Faculty of Pharmacy, University of Granada, Granada, Spain
- Instituto de Nutrición y Tecnología de los Alimentos, Biomedical Research Centre, University of Granada, Granada, Spain
| | - Mercedes G Bermúdez
- Department of Paediatrics, Faculty of Medicine, University of Granada, Granada, Spain;
- Instituto de Investigación Biosanitaria (IBS-GRANADA), Granada, Spain
- Instituto de Neurociencias "Doctor Federico Olóriz," Biomedical Research Centre, University of Granada, Granada, Spain
| | - Cristina Campoy
- Department of Paediatrics, Faculty of Medicine, University of Granada, Granada, Spain;
- Instituto de Investigación Biosanitaria (IBS-GRANADA), Granada, Spain
- Instituto de Neurociencias "Doctor Federico Olóriz," Biomedical Research Centre, University of Granada, Granada, Spain
- Spanish Network of Biomedical Research in Epidemiology and Public Health, Granada Node, Carlos III Health Institute, Madrid, Spain
| |
Collapse
|
15
|
Mallott EK, Sitarik AR, Leve LD, Cioffi C, Camargo CA, Hasegawa K, Bordenstein SR. Human microbiome variation associated with race and ethnicity emerges as early as 3 months of age. PLoS Biol 2023; 21:e3002230. [PMID: 37590208 PMCID: PMC10434942 DOI: 10.1371/journal.pbio.3002230] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 07/03/2023] [Indexed: 08/19/2023] Open
Abstract
Human microbiome variation is linked to the incidence, prevalence, and mortality of many diseases and associates with race and ethnicity in the United States. However, the age at which microbiome variability emerges between these groups remains a central gap in knowledge. Here, we identify that gut microbiome variation associated with race and ethnicity arises after 3 months of age and persists through childhood. One-third of the bacterial taxa that vary across caregiver-identified racial categories in children are taxa reported to also vary between adults. Machine learning modeling of childhood microbiomes from 8 cohort studies (2,756 samples from 729 children) distinguishes racial and ethnic categories with 87% accuracy. Importantly, predictive genera are also among the top 30 most important taxa when childhood microbiomes are used to predict adult self-identified race and ethnicity. Our results highlight a critical developmental window at or shortly after 3 months of age when social and environmental factors drive race and ethnicity-associated microbiome variation and may contribute to adult health and health disparities.
Collapse
Affiliation(s)
- Elizabeth K. Mallott
- Vanderbilt Microbiome Innovation Center, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Biology, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Alexandra R. Sitarik
- Department of Public Health Sciences, Henry Ford Health, Detroit, Michigan, United States of America
| | - Leslie D. Leve
- Prevention Science Institute, University of Oregon, Eugene, Oregon, United States of America
| | - Camille Cioffi
- Prevention Science Institute, University of Oregon, Eugene, Oregon, United States of America
| | - Carlos A. Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Seth R. Bordenstein
- Vanderbilt Microbiome Innovation Center, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, School of Medicine, Nashville, Tennessee, United States of America
- Departments of Biology and Entomology, Pennsylvania State University, University Park, Pennsylvania, United States of America
- The One Health Microbiome Center, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
16
|
Luecke SM, Holman DB, Schmidt KN, Gzyl KE, Hurlbert JL, Menezes ACB, Bochantin KA, Kirsch JD, Baumgaertner F, Sedivec KK, Swanson KC, Dahlen CR, Amat S. Whole-body microbiota of newborn calves and their response to prenatal vitamin and mineral supplementation. Front Microbiol 2023; 14:1207601. [PMID: 37434710 PMCID: PMC10331429 DOI: 10.3389/fmicb.2023.1207601] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/08/2023] [Indexed: 07/13/2023] Open
Abstract
Early life microbial colonization and factors affecting colonization patterns are gaining interest due to recent developments suggesting that early life microbiome may play a role in Developmental Origins of Health and Disease. In cattle, limited information exists on the early microbial colonization of anatomical sites involved in bovine health beyond the gastrointestinal tract. Here, we investigated 1) the initial microbial colonization of seven different anatomical locations in newborn calves and 2) whether these early life microbial communities and 3) serum cytokine profiles are influenced by prenatal vitamin and mineral (VTM) supplementation. Samples were collected from the hoof, liver, lung, nasal cavity, eye, rumen (tissue and fluid), and vagina of beef calves that were born from dams that either received or did not receive VTM supplementation throughout gestation (n = 7/group). Calves were separated from dams immediately after birth and fed commercial colostrum and milk replacer until euthanasia at 30 h post-initial colostrum feeding. The microbiota of all samples was assessed using 16S rRNA gene sequencing and qPCR. Calf serum was subjected to multiplex quantification of 15 bovine cytokines and chemokines. Our results indicated that the hoof, eye, liver, lung, nasal cavity, and vagina of newborn calves were colonized by site-specific microbiota, whose community structure differed from the ruminal-associated communities (0.64 ≥ R2 ≥ 0.12, p ≤ 0.003). The ruminal fluid microbial community was the only one that differed by treatment (p < 0.01). However, differences (p < 0.05) by treatment were detected in microbial richness (vagina); diversity (ruminal tissue, fluid, and eye); composition at the phylum and genus level (ruminal tissue, fluid, and vagina); and in total bacterial abundance (eye and vagina). From serum cytokines evaluated, concentration of chemokine IP-10 was greater (p = 0.02) in VTM calves compared to control calves. Overall, our results suggest that upon birth, the whole-body of newborn calves are colonized by relatively rich, diverse, and site-specific bacterial communities. Noticeable differences were observed in ruminal, vaginal, and ocular microbiota of newborn calves in response to prenatal VTM supplementation. These findings can derive future hypotheses regarding the initial microbial colonization of different body sites, and on maternal micronutrient consumption as a factor that may influence early life microbial colonization.
Collapse
Affiliation(s)
- Sarah M. Luecke
- Department of Microbiological Sciences, North Dakota State University, Fargo, ND, United States
| | - Devin B. Holman
- Lacombe Research and Development Centre, Agriculture and Agri-Food Canada, Lacombe, AB, Canada
| | - Kaycie N. Schmidt
- Department of Microbiological Sciences, North Dakota State University, Fargo, ND, United States
| | - Katherine E. Gzyl
- Lacombe Research and Development Centre, Agriculture and Agri-Food Canada, Lacombe, AB, Canada
| | - Jennifer L. Hurlbert
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Ana Clara B. Menezes
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Kerri A. Bochantin
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - James D. Kirsch
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Friederike Baumgaertner
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Kevin K. Sedivec
- Central Grasslands Research Extension Center, North Dakota State University, Streeter, ND, United States
| | - Kendall C. Swanson
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Carl R. Dahlen
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Samat Amat
- Department of Microbiological Sciences, North Dakota State University, Fargo, ND, United States
| |
Collapse
|
17
|
Avizemel O, Frishman S, Pinto Y, Michael Y, Turjeman S, Tenenbaum-Gavish K, Yariv O, Peled Y, Poran E, Pardo J, Chen R, Hod M, Schwartz B, Hadar E, Koren O, Agay-Shay K. "Residential greenness, gestational diabetes mellitus (GDM) and microbiome diversity during pregnancy". Int J Hyg Environ Health 2023; 251:114191. [PMID: 37290331 DOI: 10.1016/j.ijheh.2023.114191] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 04/11/2023] [Accepted: 05/22/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is associated with reduced gut microbiota richness that was also reported to differ significantly between those living in rural compared to urban environments. Therefore, our aim was to examine the associations between greenness and maternal blood glucose levels and GDM, with microbiome diversity as a possible mediator in these associations. METHODS Pregnant women were recruited between January 2016 and October 2017. Residential greenness was evaluated as mean Normalized Difference Vegetation Index (NDVI) within 100, 300 and 500 m buffers surrounding each maternal residential address. Maternal glucose levels were measured at 24-28 weeks of gestation and GDM was diagnosed. We estimated the associations between greenness and glucose levels and GDM using generalized linear models, adjusting for socioeconomic status and season at last menstrual period. Using causal mediation analysis, the mediation effects of four different indices of microbiome alpha diversity in first trimester stool and saliva samples were assessed. RESULTS Of 269 pregnant women, 27 participants (10.04%) were diagnosed with GDM. Although not statistically significant, adjusted exposure to medium tertile levels of mean NDVI at 300 m buffer had lower odds of GDM (OR = 0.45, 95% CI: 0.16, 1.26, p = 0.13) and decreased change in mean glucose levels (β = -6.28, 95% CI: 14.91, 2.24, p = 0.15) compared to the lowest tertile levels of mean NDVI. Mixed results were observed at 100 and 500 m buffers, and when comparing highest tertile levels to lowest. No mediation effect of first trimester microbiome on the association between residential greenness and GDM was observed, and a small, possibly incidental, mediation effect on glucose levels was observed. CONCLUSION Our study suggests possible associations between residential greenness and glucose intolerance and risk of GDM, though without sufficient evidence. Microbiome in the first trimester, while involved in GDM etiology, is not a mediator in these associations. Future studies in larger populations should further examine these associations.
Collapse
Affiliation(s)
- Ofir Avizemel
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel; The Health & Environment Research (HER) Lab, Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel.
| | - Sigal Frishman
- Institute of Biochemistry, School of Nutritional Sciences Food Science and Nutrition, The School of Nutritional Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel; Helen Schneider Hospital for Women, Rabin Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | - Yishay Pinto
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Yaron Michael
- Department of Soil & Water Sciences, Institute of Environmental Sciences, the Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Sondra Turjeman
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Kinneret Tenenbaum-Gavish
- Helen Schneider Hospital for Women, Rabin Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | - Or Yariv
- Department of Soil & Water Sciences, Institute of Environmental Sciences, the Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Yoav Peled
- Helen Schneider Hospital for Women, Rabin Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Israel; Clalit Medical Services, Dan Petach-Tikva District, Israel
| | - Eran Poran
- Clalit Medical Services, Dan Petach-Tikva District, Israel
| | - Joseph Pardo
- Helen Schneider Hospital for Women, Rabin Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Israel; Clalit Medical Services, Dan Petach-Tikva District, Israel
| | - Rony Chen
- Helen Schneider Hospital for Women, Rabin Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | - Moshe Hod
- Helen Schneider Hospital for Women, Rabin Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | - Betty Schwartz
- Institute of Biochemistry, School of Nutritional Sciences Food Science and Nutrition, The School of Nutritional Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Eran Hadar
- Helen Schneider Hospital for Women, Rabin Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Keren Agay-Shay
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel; The Health & Environment Research (HER) Lab, Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel.
| |
Collapse
|
18
|
Mady EA, Doghish AS, El-Dakroury WA, Elkhawaga SY, Ismail A, El-Mahdy HA, Elsakka EGE, El-Husseiny HM. Impact of the mother's gut microbiota on infant microbiome and brain development. Neurosci Biobehav Rev 2023; 150:105195. [PMID: 37100161 DOI: 10.1016/j.neubiorev.2023.105195] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/18/2023] [Accepted: 04/21/2023] [Indexed: 04/28/2023]
Abstract
The link between the gut microbiome and health has recently garnered considerable interest in its employment for medicinal purposes. Since the early microbiota exhibits more flexibility compared to that of adults, there is a considerable possibility that altering it will have significant consequences on human development. Like genetics, the human microbiota can be passed from mother to child. This provides information on early microbiota acquisition, future development, and prospective chances for intervention. The succession and acquisition of early-life microbiota, modifications of the maternal microbiota during pregnancy, delivery, and infancy, and new efforts to understand maternal-infant microbiota transmission are discussed in this article. We also examine the shaping of mother-to-infant microbial transmission, and we then explore possible paths for future research to advance our knowledge in this area.
Collapse
Affiliation(s)
- Eman A Mady
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan; Department of Animal Hygiene, Behavior and Management, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya,13736, Egypt.
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and industrial pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Samy Y Elkhawaga
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| | - Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Hussein M El-Husseiny
- Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan.
| |
Collapse
|
19
|
Rodrigues EL, Figueiredo PS, Marcelino G, de Cássia Avellaneda Guimarães R, Pott A, Santana LF, Hiane PA, do Nascimento VA, Bogo D, de Cássia Freitas K. Maternal Intake of Polyunsaturated Fatty Acids in Autism Spectrum Etiology and Its Relation to the Gut Microbiota: What Do We Know? Nutrients 2023; 15:nu15071551. [PMID: 37049390 PMCID: PMC10097097 DOI: 10.3390/nu15071551] [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: 01/17/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 04/14/2023] Open
Abstract
Maternal food habits and gut microbiota composition have potential effects on fetal neurodevelopment, impacting Autism Spectrum Disorder (ASD). Our research aims to outline the relationship that ingestion of polyunsaturated fatty acids (PUFAs) and the composition of maternal gut microbiota have with the possible development of ASD in offspring. We suggest that genetic factors could be related to the different conversions between unsaturated fatty acids according to sex and, mainly, the impact of the pregnancy diet on the higher or lower risk of neurological impairments. The proportion of the phyla Firmicutes/Bacteroidetes is high with an increased consumption of linoleic acid (LA, n-6 PUFA), which is associated with maternal intestinal dysbiosis and consequently starts the inflammatory process, harming myelinization. In contrast, the consumption of α-linolenic acid (ALA, n-3 PUFA) tends to re-establish the balance of the maternal microbiota with anti-inflammatory action. Moreover, human observational studies showed a strong correlation between the consumption of n-3 PUFA, mainly above 340 g of fish per week, with beneficial effects on infant neurodevelopment. Therefore, we suggest that the proper intake of foods rich in n-3 PUFAs and their supplementation during pregnancy until lactation has an impact on reducing the development of ASD. Controlled studies with n-3 PUFA supplementation are still necessary to verify the ideal dose and the best form of administration.
Collapse
Affiliation(s)
- Elisana Lima Rodrigues
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Priscila Silva Figueiredo
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Gabriela Marcelino
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Rita de Cássia Avellaneda Guimarães
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Arnildo Pott
- Institute of Biosciences, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Lidiani Figueiredo Santana
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Priscila Aiko Hiane
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Valter Aragão do Nascimento
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Danielle Bogo
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Karine de Cássia Freitas
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| |
Collapse
|
20
|
Tian M, Li Q, Zheng T, Yang S, Chen F, Guan W, Zhang S. Maternal microbe-specific modulation of the offspring microbiome and development during pregnancy and lactation. Gut Microbes 2023; 15:2206505. [PMID: 37184203 DOI: 10.1080/19490976.2023.2206505] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
The maternal microbiome is essential for the healthy growth and development of offspring and has long-term effects later in life. Recent advances indicate that the maternal microbiome begins to regulate fetal health and development during pregnancy. Furthermore, the maternal microbiome continues to affect early microbial colonization via birth and breastfeeding. Compelling evidence indicates that the maternal microbiome is involved in the regulation of immune and brain development and affects the risk of related diseases. Modulating offspring development by maternal diet and probiotic intervention during pregnancy and breastfeeding could be a promising therapy in the future. In this review, we summarize and discuss the current understanding of maternal microbiota development, perinatal microbial metabolite transfer, mother-to-infant microbial transmission during/after birth and its association with immune and brain development as well as corresponding diseases.
Collapse
Affiliation(s)
- Min Tian
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Tenghui Zheng
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Fang Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
21
|
Shi F, Almerick T Boncan D, Wan HT, Chan TF, Zhang EL, Lai KP, Wong CKC. Hepatic metabolism gene expression and gut microbes in offspring, subjected to in-utero PFOS exposure and postnatal diet challenges. CHEMOSPHERE 2022; 308:136196. [PMID: 36041519 DOI: 10.1016/j.chemosphere.2022.136196] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 06/15/2023]
Abstract
We examined the changes in hepatic metabolic gene expression and gut microbiota of offspring exposed to PFOS in-utero. At GD17.5, our data showed that PFOS exposure decreased fetal bodyweights and hepatic metabolic gene expressions but increased relative liver mass and lipid accumulation. At PND21, in-utero high-dose PFOS-exposed offspring exhibited significantly greater bodyweight (catch-up-growth), associated with significant induction of hepatic metabolic gene expression. In addition, 16SrRNA-sequencing of the cecal samples revealed an increase in carbohydrate catabolism but a reduction in microbial polysaccharide synthesis and short-chain fatty acid (SCFA) metabolism. From PND21-80, a postnatal diet-challenge for the offspring was conducted. At PND80 under a normal diet, in-utero high-dose PFOS-exposed offspring maintained the growth "catch-up" effect. In contrast, in a high-fat-diet, the bodyweight of in-utero high-dose PFOS-exposed adult offspring were significantly lesser than the corresponding low-dose and control groups. Even though in the high-fat-diet, the in-utero PFOS-exposed adult offspring showed significant upregulation of hepatic metabolic genes, the lower bodyweight suggests that they had difficulty utilizing high-fat nutrients. Noteworthy, the metagenomic data showed a significant reduction in the biosynthesis of microbial polysaccharides, vitamin B, and SCFAs in the PFOS-exposed adult offspring. Furthermore, the observed effects were significantly reduced in the PFOS-exposed adult offspring with the high-fat diet but supplemented with sucrose. Our study demonstrated that in-utero PFOS exposure caused inefficient fat metabolism and increased the risk of hepatic steatosis in offspring.
Collapse
Affiliation(s)
- Feng Shi
- State Key Laboratory in Environmental and Biological Analysis, Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China
| | - Delbert Almerick T Boncan
- School of Life Sciences, State Key Laboratory of Agrobiotechnology, Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hin Ting Wan
- State Key Laboratory in Environmental and Biological Analysis, Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ting Fung Chan
- School of Life Sciences, State Key Laboratory of Agrobiotechnology, Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Eric L Zhang
- Department of Computer Science, Hong Kong Baptist University, Hong Kong SAR, China
| | - Keng Po Lai
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR China
| | - Chris Kong-Chu Wong
- State Key Laboratory in Environmental and Biological Analysis, Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
22
|
Zeng S, Wang S, Ross RP, Stanton C. The road not taken: host genetics in shaping intergenerational microbiomes. Trends Genet 2022; 38:1180-1192. [PMID: 35773025 DOI: 10.1016/j.tig.2022.05.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/31/2022] [Accepted: 05/31/2022] [Indexed: 02/09/2023]
Abstract
The early-life gut microbiome is linked to human phenotypes as an imbalanced microbiome of this period is implicated in diseases throughout life. Several determinants of early-life gut microbiome are explored, however, mechanisms of acquisition, colonization, and stability of early-life gut microbiome and their interindividual variability remain elusive. Host genetics play a vital role to shape the gut microbiome and interact with it to modulate individual phenotypes in human studies and animal models. Given the microbial linkage between host generations, we discuss the current state of roles of host genetics in forming intergenerational microbiomes associated with mothers, offspring, and those vertically transmitted, providing a basis for taking into account host genetics in future early-life microbiome research. We further expand our discussion to the bidirectional interactions between host gene expression and microbiome in human health.
Collapse
Affiliation(s)
- Shuqin Zeng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China; APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, P61 C996, Ireland
| | - Shaopu Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China; APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, P61 C996, Ireland.
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, P61 C996, Ireland
| |
Collapse
|
23
|
Berti C, Elahi S, Catalano P, Bhutta ZA, Krawinkel MB, Parisi F, Agostoni C, Cetin I, Hanson M. Obesity, Pregnancy and the Social Contract with Today's Adolescents. Nutrients 2022; 14:3550. [PMID: 36079808 PMCID: PMC9459961 DOI: 10.3390/nu14173550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 12/16/2022] Open
Abstract
Adolescent health and well-being are of great concern worldwide, and adolescents encounter particular challenges, vulnerabilities and constraints. The dual challenges of adolescent parenthood and obesity are of public health relevance because of the life-altering health and socioeconomic effects on both the parents and the offspring. Prevention and treatment strategies at the individual and population levels have not been successful in the long term, suggesting that adolescent pregnancy and obesity cannot be managed by more of the same. Here, we view adolescent obese pregnancy through the lens of the social contract with youth. The disruption of this contract is faced by today's adolescents, with work, social and economic dilemmas which perpetuate socioeconomic and health inequities across generations. The lack of employment, education and social opportunities, together with obesogenic settings, increase vulnerability and exposure to lifelong health risks, affecting their offspring's life chances too. To break such vicious circles of disadvantage and achieve sustainable solutions in real-world settings, strong efforts on the part of policymakers, healthcare providers and the community must be oriented towards guaranteeing equity and healthy nutrition and environments for today's adolescents. The involvement of adolescents themselves in developing such programs is paramount, not only so that they feel a sense of agency but also to better meet their real life needs.
Collapse
Affiliation(s)
- Cristiana Berti
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pediatric Unit, 20122 Milan, Italy
| | | | - Patrick Catalano
- Mother Infant Research Institute, Tufts University School of Medicine, Boston 02111, MA, USA
| | - Zulfiqar A. Bhutta
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Center of Excellence in Women and Child Health, The Aga Khan University, Karachi 74800, Pakistan
| | - Michael B. Krawinkel
- Institute of Nutritional Sciences—International Nutrition, Justus-Liebig-University, 35392 Giessen, Germany
| | - Francesca Parisi
- Department of Woman, Mother and Neonate, “V. Buzzi” Children Hospital, ASST Fatebenefratelli Sacco, 20154 Milan, Italy
| | - Carlo Agostoni
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pediatric Unit, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Irene Cetin
- Department of Woman, Mother and Neonate, “V. Buzzi” Children Hospital, ASST Fatebenefratelli Sacco, 20154 Milan, Italy
- Department of Biomedical and Clinical Sciences, School of Medicine, University of Milan, 20154 Milan, Italy
| | - Mark Hanson
- Institute of Developmental Sciences and NIHR Biomedical Research Centre, University of Southampton and University Hospital Southampton, Southampton SO17 1BJ, UK
| |
Collapse
|
24
|
Zheng J, Zhang L, Gao Y, Wu H, Zhang J. The dynamic effects of maternal high-calorie diet on glycolipid metabolism and gut microbiota from weaning to adulthood in offspring mice. Front Nutr 2022; 9:941969. [PMID: 35928844 PMCID: PMC9343994 DOI: 10.3389/fnut.2022.941969] [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: 05/12/2022] [Accepted: 07/11/2022] [Indexed: 12/04/2022] Open
Abstract
Dysbiosis of gut microbiota can contribute to the progression of diabetes and obesity. Previous studies have shown that maternal high-fat (HF) diet during the perinatal period can alter the microbiota and induce metabolic disorders at weaning. However, whether dysbiosis of gut microbiota and metabolism could be recovered by a normal diet after weaning and the dynamic changes of gut microbiota have not been fully studied. In this study, C57BL/6J female mice were fed with a normal chow (NC) or HF diet for 4 weeks preconception, during gestation, and until pup weaning. After weaning, male offspring were fed with an NC diet until 9 weeks of age. The microbiota of offspring at weaning and 9 weeks of age was collected for 16S rRNA gene amplicon sequencing. We found that dams fed with an HF diet showed glucose intolerance after lactation. Compared with the offspring from NC dams, the offspring from HF dams exhibited a higher body weight, hyperglycemia, glucose intolerance, hyperinsulinemia, hypercholesterolemia, and leptin resistance and lower adiponectin at weaning. Fecal analysis indicated altered microbiota composition between the offspring of the two groups. The decrease in favorable bacteria (such as norank f Bacteroidales S24-7 group) and increase in unfavorable bacteria (such as Lachnoclostridium and Desulfovibrio) were strongly associated with a disturbance of glucose and lipid metabolism. After 6 weeks of normal diet, no difference in body weight, glucose, and lipid profiles was observed between the offspring of the two groups. However, the microbiota composition of offspring in the HF group was still different from that in the NC group, and microbiota diversity was lower in offspring of the HF group. The abundance of Lactobacillus was lower in the offspring of the HF group. In conclusion, a maternal HF diet can induce metabolic homeostasis and gut microbiota disturbance in offspring at weaning. Gut microbiota dysbiosis can persist into adulthood in the offspring, which might have a role in the promotion of susceptibility to obesity and diabetes in the later life of the offspring.
Collapse
Affiliation(s)
- Jia Zheng
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Ling Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Ying Gao
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Honghua Wu
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Junqing Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| |
Collapse
|
25
|
Vuong HE. Intersections of the microbiome and early neurodevelopment. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 167:1-23. [PMID: 36427952 DOI: 10.1016/bs.irn.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Our resident microbes influence nearly all aspects of our biological systems. In particular, the maternal and early life microbiota is uniquely positioned to influence the development of the nervous system, and alterations to the gut microbiota, or dysbiosis, during this critical time in early life can have long-lasting negative effects on health. The question of how the maternal and early life microbiota shapes neurodevelopment is the topic of numerous investigations. Here, we discuss two possible, but not necessarily independent, hypotheses: (1) the maternal microbiota during pregnancy regulates the metabolites that are important for fetal development, (2) maternal microbiota seeded to offspring at birth and early postnatal days programs offspring immune and brain development, and regulates key molecules for postnatal brain development. In this chapter, we provide an overview of the impact of the microbiota on brain and behavior, introduce the maternal gut and vaginal microbiome during pregnancy, and discuss current understandings of microbiome in the context of developmental origins of health and disease. We consider novel translational insights that harness the multitude of microbes and microbial metabolites for prevention or treatment of neurological disorders.
Collapse
Affiliation(s)
- Helen E Vuong
- Department of Pediatrics, Division of Neonatology, University of Minnesota Twin Cities, Minneapolis, MN, United States.
| |
Collapse
|
26
|
Koemel NA, Skilton MR. Epigenetic Aging in Early Life: Role of Maternal and Early Childhood Nutrition. Curr Nutr Rep 2022; 11:318-328. [PMID: 35192186 PMCID: PMC9174131 DOI: 10.1007/s13668-022-00402-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2022] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Early life presents a pivotal period during which nutritional exposures are more likely to cause epigenetic modifications, which may impact an individual's health during adulthood. This article reviews the current evidence regarding maternal and early childhood nutritional exposures and their role in epigenetic aging. RECENT FINDINGS Maternal and early life consumption of diets higher in fiber, antioxidants, polyphenols, B vitamins, vitamin D, and ω-3 fatty acids is associated with slower epigenetic aging. Conversely, diets higher in glycemic load, fat, saturated fat, and ω-6 fatty acids demonstrate a positive association with epigenetic aging. Maternal and early life nutrition directly and indirectly influences epigenetic aging via changes in one-carbon metabolism, cardiometabolic health, and the microbiome. Clinical trials are warranted to determine the specific foods, dietary patterns, and dietary supplements that will normalize or lower epigenetic aging across the life course.
Collapse
Affiliation(s)
- Nicholas A. Koemel
- The Boden Initiative, Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Michael R. Skilton
- The Boden Initiative, Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Sydney Institute for Women, Children and Their Families, Sydney Local Health District, Sydney, Australia
| |
Collapse
|
27
|
Hasebe K, Kendig MD, Kaakoush NO, Tajaddini A, Westbrook RF, Morris MJ. The influence of maternal unhealthy diet on maturation of offspring gut microbiota in rat. Anim Microbiome 2022; 4:31. [PMID: 35551670 PMCID: PMC9102338 DOI: 10.1186/s42523-022-00185-w] [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: 10/21/2021] [Accepted: 04/27/2022] [Indexed: 12/04/2022] Open
Abstract
Background Despite well-known effects of diet on gut microbiota diversity, relatively little is known about how maternal diet quality shapes the longitudinal maturation of gut microbiota in offspring. To investigate, we fed female rats standard chow (Chow) or a western-style, high-choice cafeteria diet (Caf) prior to and during mating, gestation and lactation. At weaning (3 weeks), male and female offspring were either maintained on their mother’s diet (ChowChow, CafCaf groups) or switched to the other diet (ChowCaf, CafChow). Fecal microbial composition was assessed in dams and longitudinally in offspring at 3, 7 and 14 weeks of age. Results The effect of maternal diet on maturation of offspring gut microbiota was assessed by α- and β-diversities, Deseq2/LEfSe, and SourceTracker analyses. Weanling gut microbiota composition was characterised by reduced α- and β-diversity profiles that clustered away from dams and older siblings. After weaning, offspring gut microbiota came to resemble an adult-like gut microbiota, with increased α-diversity and reduced dissimilarity of β-diversity. Similarly, Deseq2/LEfSe analyses found fewer numbers of altered operational taxonomic units (OTUs) between groups from weaning to adulthood. SourceTracker analyses indicated a greater overall contribution of Caf mothers’ microbial community (up to 20%) to that of their offspring than the contribution of Chow mothers (up to 8%). Groups maintained on the maternal diet (ChowChow, CafCaf), versus those switched to the other diet (ChowCaf, CafChow) post-weaning significantly differed from each other at 14 weeks (Permutational Multivariate Analysis of Variance), indicating interactive effects of maternal and post-weaning diet on offspring gut microbiota maturation. Nevertheless, this developmental trajectory was unaffected by sex and appeared consistent between ChowChow, CafCaf, ChowCaf and CafChow groups. Conclusions Introducing solid food at weaning triggered the maturation of offspring gut microbiota to an adult-like profile in rats, in line with previous human studies. Postweaning Caf diet exposure had the largest impact on offspring gut microbiota, but this was modulated by maternal diet history. An unhealthy maternal Caf diet did not alter the developmental trajectory of offspring gut microbiota towards an adult-like profile, insofar as it did not prevent the age-associated increase in α-diversity and reduction in β-diversity dissimilarity. Supplementary Information The online version contains supplementary material available at 10.1186/s42523-022-00185-w.
Collapse
Affiliation(s)
- Kyoko Hasebe
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Michael D Kendig
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Nadeem O Kaakoush
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Aynaz Tajaddini
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, 2052, Australia
| | | | - Margaret J Morris
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, 2052, Australia.
| |
Collapse
|
28
|
Amat S, Dahlen CR, Swanson KC, Ward AK, Reynolds LP, Caton JS. Bovine Animal Model for Studying the Maternal Microbiome, in utero Microbial Colonization and Their Role in Offspring Development and Fetal Programming. Front Microbiol 2022; 13:854453. [PMID: 35283808 PMCID: PMC8916045 DOI: 10.3389/fmicb.2022.854453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/07/2022] [Indexed: 01/10/2023] Open
Abstract
Recent developments call for further research on the timing and mechanisms involved in the initial colonization of the fetal/infant gut by the maternal microbiome and its role in Developmental Origins of Health and Disease (DOHaD). Although progress has been made using primarily preterm infants, ethical and legal constraints hinder research progress in embryo/fetal-related research and understanding the developmental and mechanistic roles of the maternal microbiome in fetal microbial imprinting and its long-term role in early-life microbiome development. Rodent models have proven very good for studying the role of the maternal microbiome in fetal programming. However, some inherent limitations in these animal models make it challenging to study perinatal microbial colonization from a biomedical standpoint. In this review, we discuss the potential use of bovine animals as a biomedical model to study the maternal microbiome, in utero microbial colonization of the fetal gut, and their impact on offspring development and DOHaD.
Collapse
Affiliation(s)
- Samat Amat
- Department of Microbiological Sciences, North Dakota State University, Fargo, ND, United States
| | - Carl R Dahlen
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Kendall C Swanson
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Alison K Ward
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Lawrence P Reynolds
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Joel S Caton
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| |
Collapse
|
29
|
Amat S, Holman DB, Schmidt K, McCarthy KL, Dorsam ST, Ward AK, Borowicz PP, Reynolds LP, Caton JS, Sedivec KK, Dahlen CR. Characterization of the Microbiota Associated With 12-Week-Old Bovine Fetuses Exposed to Divergent in utero Nutrition. Front Microbiol 2022; 12:771832. [PMID: 35126326 PMCID: PMC8811194 DOI: 10.3389/fmicb.2021.771832] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/24/2021] [Indexed: 12/18/2022] Open
Abstract
A recent study reported the existence of a diverse microbiota in 5-to-7-month-old calf fetuses, suggesting that colonization of the bovine gut with so-called “pioneer” microbiota may begin during mid-gestation. In the present study, we investigated 1) the presence of microbiota in bovine fetuses at early gestation (12 weeks), and 2) whether the fetal microbiota is influenced by the maternal rate of gain or dietary supplementation with vitamins and minerals (VTM) during early gestation. Amniotic and allantoic fluids, and intestinal and placental (cotyledon) tissue samples obtained from fetuses (n = 33) on day 83 of gestation were processed for the assessment of fetal microbiota using 16S rRNA gene sequencing. The sequencing results revealed that a diverse and complex microbial community was present in each of these fetal compartments evaluated. Allantoic and amniotic fluids, and fetal intestinal and placenta microbiota each had distinctly different (0.047 ≥ R2 ≥ 0.019, P ≤ 0.031) microbial community structures. Allantoic fluid had a greater (P < 0.05) microbial richness (number of OTUs) (Mean 122) compared to amniotic fluid (84), intestine (63), and placenta (66). Microbial diversity (Shannon index) was similar for the intestinal and placental samples, and both were less diverse compared with fetal fluid microbiota (P < 0.05). Thirty-nine different archaeal and bacterial phyla were detected across all fetal samples, with Proteobacteria (55%), Firmicutes (16.2%), Acidobacteriota (13.6%), and Bacteroidota (5%) predominating. Among the 20 most relatively abundant bacterial genera, Acidovorax, Acinetobacter, Brucella, Corynebacterium, Enterococcus, Exiguobacterium, and Stenotrophomonas differed by fetal sample type (P < 0.05). A total of 55 taxa were shared among the four different microbial communities. qPCR of bacteria in the intestine and placenta samples as well as scanning electron microscopy imaging of fetal fluids provided additional evidence for the presence of a microbiota in these samples. Minor effects of maternal rate of gain and VTM supplementation, and their interactions on microbial richness and composition were detected. Overall, the results of this study indicate that colonization with pioneer microbiota may occur during early gestation in bovine fetuses, and that the maternal nutritional regime during gestation may influence the early fetal microbiota.
Collapse
Affiliation(s)
- Samat Amat
- Department of Microbiological Sciences, North Dakota State University, Fargo, ND, United States
- *Correspondence: Samat Amat,
| | - Devin B. Holman
- Lacombe Research and Development Centre, Agriculture and Agri-Food Canada, Lacombe, AB, Canada
| | - Kaycie Schmidt
- Department of Microbiological Sciences, North Dakota State University, Fargo, ND, United States
| | - Kacie L. McCarthy
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Sheri T. Dorsam
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Alison K. Ward
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Pawel P. Borowicz
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Lawrence P. Reynolds
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Joel S. Caton
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Kevin K. Sedivec
- Central Grasslands Research Extension Center, North Dakota State University, Streeter, ND, United States
| | - Carl R. Dahlen
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| |
Collapse
|
30
|
Huang H, Peng Q, Zhang Y, Li Y, Huang N, Duan M, Huang B. Abnormalities in microbial composition and function in infants with necrotizing enterocolitis: A single-center observational study. Front Pediatr 2022; 10:963345. [PMID: 36340725 PMCID: PMC9634528 DOI: 10.3389/fped.2022.963345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To investigate the features and functions of the intestinal microbiota in neonates with necrotizing enterocolitis (NEC) in a single center in China. METHODS We collected clinical information and stool samples from 19 participants in our center, including 9 infants with necrotizing enterocolitis and 10 control infants. DNA was extracted from the samples, and 16S rRNA gene sequencing was used to analyse the participants' gut microbiota. Functional prediction was achieved using PICRUSt2. RESULTS Alpha diversity analysis found that similar levels of bacterial richness and diversity were found in the gut microbiota of infants with NEC and control infants (P = 0.1800), whereas beta diversity analysis suggested that the overall structures of the gut microbiota were significantly different (P = 0.0020). The Mann-Whitney U test of bacterial composition and abundance analysis revealed that the abundance levels of Proteobacteria (P = 0.03049) and Firmicutes (P = 0.01011) significantly differed between the two groups at the phylum level. Proteobacteria was the most abundant phylum in the NEC group. At the genus level, the abundance levels of Enterococcus (P = 0.0003), Streptococcaceae (P = 0.0109) and Lactobacillales (P = 0.0171) were significantly decreased in infants with NEC. Furthermore, the linear discriminant analysis effect size (LEfSe) method showed 12 bacterial taxa with significant differences in relative abundances in the two groups. Interestingly, members of Proteobacteria were enriched in NEC samples. In addition, functional prediction suggested that the microbial changes observed in infants with NEC resulted in a decline in galactose metabolism, the pentose phosphate pathway, fructose and mannose metabolism, amino sugar and nucleotide sugar metabolism, glycolysis/gluconeogenesis, starch and sucrose metabolism, and phosphotransferase system (PTS) pathways (P < 0.05). CONCLUSIONS Our study shows the compositional and functional alterations of the intestinal microbiota in NEC, which will help demonstrate the relationship between the gut microbiota and NEC pathogenesis.
Collapse
Affiliation(s)
- Huan Huang
- Department of Pediatrics, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, China
| | - Qian Peng
- Department of Pediatrics, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, China
| | - Yuli Zhang
- Department of Pediatrics, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, China
| | - Ying Li
- Department of Pediatrics, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, China
| | - Nanqu Huang
- National Drug Clinical Trial Institution, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, China
| | - Miao Duan
- Department of Pediatrics, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, China
| | - Bo Huang
- Department of Pediatrics, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, China
| |
Collapse
|
31
|
Healy DB, Ryan CA, Ross RP, Stanton C, Dempsey EM. Clinical implications of preterm infant gut microbiome development. Nat Microbiol 2022; 7:22-33. [PMID: 34949830 DOI: 10.1038/s41564-021-01025-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022]
Abstract
Perturbations to the infant gut microbiome during the first weeks to months of life affect growth, development and health. In particular, assembly of an altered intestinal microbiota during infant development results in an increased risk of immune and metabolic diseases that can persist into childhood and potentially into adulthood. Most research into gut microbiome development has focused on full-term babies, but health-related outcomes are also important for preterm babies. The systemic physiological immaturity of very preterm gestation babies (born earlier than 32 weeks gestation) results in numerous other microbiome-organ interactions, the mechanisms of which have yet to be fully elucidated or in some cases even considered. In this Perspective, we compare assembly of the intestinal microbiome in preterm and term infants. We focus in particular on the clinical implications of preterm infant gut microbiome composition and discuss the prospects for microbiome diagnostics and interventions to improve the health of preterm babies.
Collapse
Affiliation(s)
- David B Healy
- APC Microbiome Ireland, University College Cork, Cork, Ireland. .,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.
| | - C Anthony Ryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Eugene M Dempsey
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.,INFANT Research Centre, Cork University Hospital, Cork, Ireland
| |
Collapse
|
32
|
Šeda O. Parental overnutrition by carbohydrates in developmental origins of metabolic syndrome. Physiol Res 2021; 70:S585-S596. [DOI: 10.33549/physiolres.934806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
interplay of genomic component and the exposome. Parental diet has been shown to affect offspring metabolic health via multiple epigenetic mechanisms. Excess carbohydrate intake is one of the driving forces of the obesity and metabolic syndrome pandemics. This review summarizes the evidence for the effects of maternal carbohydrate (fructose, sucrose, glucose) overnutrition on the modulation of metabolic syndrome components in the offspring. Despite substantial discrepancies in experimental design, common effects of maternal carbohydrate overnutrition include increased body weight and hepatic lipid content of the "programmed" offspring. However, the administration of sucrose to several rat models leads to apparently favorable metabolic outcomes. Moreover, there is evidence for the role of genomic background in modulating the metabolic programming effect in the form of nutri-epigenomic interaction. Comprehensive, robust studies are needed to resolve the temporal, sex-specific, genetic, epigenetic and nutritional aspects of parental overnutrition in the intergenerational and transgenerational pathogenesis of metabolic syndrome.
Collapse
|
33
|
Šeda O. Parental overnutrition by carbohydrates in developmental origins of metabolic syndrome. Physiol Res 2021. [DOI: 10.33549//physiolres.934806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Metabolic syndrome is a prevalent disease resulting from an interplay of genomic component and the exposome. Parental diet has been shown to affect offspring metabolic health via multiple epigenetic mechanisms. Excess carbohydrate intake is one of the driving forces of the obesity and metabolic syndrome pandemics. This review summarizes the evidence for the effects of maternal carbohydrate (fructose, sucrose, glucose) overnutrition on the modulation of metabolic syndrome components in the offspring. Despite substantial discrepancies in experimental design, common effects of maternal carbohydrate overnutrition include increased body weight and hepatic lipid content of the "programmed" offspring. However, the administration of sucrose to several rat models leads to apparently favorable metabolic outcomes. Moreover, there is evidence for the role of genomic background in modulating the metabolic programming effect in the form of nutri-epigenomic interaction. Comprehensive, robust studies are needed to resolve the temporal, sex-specific, genetic, epigenetic and nutritional aspects of parental overnutrition in the intergenerational and transgenerational pathogenesis of metabolic syndrome.
Collapse
Affiliation(s)
- O Šeda
- Institute of Biology and Medical Genetics, the First Faculty of Medicine, Charles University and the General University Hospital, Prague, Czech Republic
| |
Collapse
|
34
|
Geng M, Tang Y, Liu K, Huang K, Yan S, Ding P, Zhang J, Wang B, Wang S, Li S, Wu X, Cao Y, Tao F. Prenatal low-dose antibiotic exposure and children allergic diseases at 4 years of age: A prospective birth cohort study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 225:112736. [PMID: 34481356 DOI: 10.1016/j.ecoenv.2021.112736] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/27/2021] [Accepted: 08/29/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Based on a medical record or questionnaire survey approach, previous epidemiological studies have investigated associations between maternal antibiotic exposure during pregnancy and childhood allergic diseases. However, biomonitoring studies on the prenatal low-dose antibiotic exposure, mainly from the environment and contaminated food, and in relation to children allergic diseases, are missing. OBJECTIVES This research aimed to examine the associations between prenatal low-dose antibiotic exposure measured at multiple time points and children current allergic diseases at 4 years of age. METHODS The current study including 2453 mother-child pairs was based on the Ma'anshan Birth Cohort study. Selected 41 antibiotics and their two metabolites, which including human antibiotics (HAs), preferred as human antibiotics (PHAs), veterinary antibiotics (VAs) and preferred as veterinary antibiotics (PVAs), in urine samples from 2453 pregnant women were biomonitored through liquid chromatography-triple quadrupole tandem mass spectrometry. Information on children current allergic diseases were collected via validated questionnaires. Generalized estimating equation were used to explore the associations between the repeated measurements of maternal urinary antibiotic over three trimesters and current allergic diseases in children. RESULTS The detection rates of nine individual antibiotics in the three trimester during pregnancy are greater than 10%, and the 90th percentile concentration of the detected antibiotics ranges from 0.07 to 22.34 µg/g, and the 95th percentile concentration ranges from 0.17 to 59.57 µg/g. Among the participants, each one-unit concentration increment of sulfamethazine (adjusted OR=1.28, 95% CI: 1.10, 1.49, P-FDR=0.014) in the first trimester and ciprofloxacin (adjusted OR=1.17, 95% CI: 1.07, 1.28, P-FDR=0.008) in the second trimester were associated with an increased risk of current eczema in children. In the third trimester, each one-unit concentration increment of oxytetracycline (adjusted OR=1.90, 95% CI: 1.30, 2.78, P-FDR=0.014) was associated with an increased risk of current asthma in children. Gender-stratified analyses demonstrated that no gender differences were observed in the associations between prenatal antibiotic exposure and current allergic diseases in children. CONCLUSIONS Maternal exposure to certain specific VAs or PVAs (sulfamethazine, ciprofloxacin and oxytetracycline) in different trimesters was associated with an increased risk of current asthma and current eczema in 4-year-old children. No gender differences were found in these associations. Further studies are warranted to confirm our findings and explore the potential mechanisms.
Collapse
Affiliation(s)
- Menglong Geng
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Ying Tang
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Kaiyong Liu
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Kun Huang
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Shuangqin Yan
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; Ma'anshan Maternal and Child Healthcare (MCH) Center, Ma'anshan 243011, China
| | - Peng Ding
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Jingjing Zhang
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, Anhui, China
| | - Baolin Wang
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, Anhui, China
| | - Sheng Wang
- The Center for Scientific Research of Anhui Medical University, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, Anhui, China
| | - Shulong Li
- The Center for Scientific Research of Anhui Medical University, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, Anhui, China
| | - Xiaoyan Wu
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Yunxia Cao
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui, China.
| | - Fangbiao Tao
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China.
| |
Collapse
|
35
|
Svobodová J, Kreisinger J, Gvoždíková Javůrková V. Temperature-induced changes in egg white antimicrobial concentrations during pre-incubation do not influence bacterial trans-shell penetration but do affect hatchling phenotype in Mallards. PeerJ 2021; 9:e12401. [PMID: 34824913 PMCID: PMC8590799 DOI: 10.7717/peerj.12401] [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: 05/10/2021] [Accepted: 10/06/2021] [Indexed: 01/10/2023] Open
Abstract
Microbiome formation and assemblage are essential processes influencing proper embryonal and early-life development in neonates. In birds, transmission of microbes from the outer environment into the egg’s interior has been found to shape embryo viability and hatchling phenotype. However, microbial transmission may be affected by egg-white antimicrobial proteins (AMPs), whose concentration and antimicrobial action are temperature-modulated. As both partial incubation and clutch covering with nest-lining feathers during the pre-incubation period can significantly alter temperature conditions acting on eggs, we experimentally investigated the effects of these behavioural mechanisms on concentrations of both the primary and most abundant egg-white AMPs (lysozyme and avidin) using mallard (Anas platyrhychos) eggs. In addition, we assessed whether concentrations of egg-white AMPs altered the probability and intensity of bacterial trans-shell penetration, thereby affecting hatchling morphological traits in vivo. We observed higher concentrations of lysozyme in partially incubated eggs. Clutch covering with nest-lining feathers had no effect on egg-white AMP concentration and we observed no association between concentration of egg-white lysozyme and avidin with either the probability or intensity of bacterial trans-shell penetration. The higher egg-white lysozyme concentration was associated with decreased scaled body mass index of hatchlings. These outcomes demonstrate that incubation prior to clutch completion in precocial birds can alter concentrations of particular egg-white AMPs, though with no effect on bacterial transmission into the egg in vivo. Furthermore, a higher egg white lysozyme concentration compromised hatchling body condition, suggesting a potential growth-regulating role of lysozyme during embryogenesis in precocial birds.
Collapse
Affiliation(s)
- Jana Svobodová
- Faculty of Environmental Sciences, Department of Ecology, Czech University of Life Sciences, Prague, Suchdol, Czech Republic
| | - Jakub Kreisinger
- Faculty of Science, Department of Zoology, Charles University Prague, Prague, Czech Republic
| | - Veronika Gvoždíková Javůrková
- Institute of Vertebrate Biology of the Czech Academy of Sciences, Brno, Czech Republic.,Faculty of Agrobiology, Food and Natural Resources, Department of Animal Science, Czech University of Life Sciences, Prague, Suchdol, Czech Republic
| |
Collapse
|
36
|
Zhang Z, Li N, Chen R, Lee T, Gao Y, Yuan Z, Nie Y, Sun T. Prenatal stress leads to deficits in brain development, mood related behaviors and gut microbiota in offspring. Neurobiol Stress 2021; 15:100333. [PMID: 34036126 PMCID: PMC8135039 DOI: 10.1016/j.ynstr.2021.100333] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 01/15/2023] Open
Abstract
Early exposure to stressful and adverse life events at fetal and neonatal stages is one of crucial risk factors for mood disorders such as anxiety and depressive disorder in adulthood. Intergenerational effects of prenatal stress on offspring are still not fully understood. We here uncover a significant negative impact of prenatal stress on brain development in embryos and newborns, and on mood-related behaviors and gut microbiota in adult offspring. Prenatal stress leads to reduced numbers in neural progenitors and newborn neurons, and altered gene expression profiles in the mouse embryonic cerebral cortex. Adult mouse offspring exposed to prenatal stress displays altered gene expression in the cortex and elevated responses in anxiety- and depression-like behaviors. Interestingly, prenatal stress has an enduring effect on gut microbiota, as specific microbial community structure is altered in adult F1 offspring treated with prenatal stress, compared to that of the control. Our results highlight the essential impact of prenatal stress on cortical neurogenesis, gene expression patterns, mood-related behaviors, and even gut microbiota in the next generation.
Collapse
Affiliation(s)
- Zhen Zhang
- School of Life Sciences and Technology, Shanghai Jiao Tong University, Shanghai, China
| | - Na Li
- School of Life Sciences and Technology, Shanghai Jiao Tong University, Shanghai, China
| | - Renliang Chen
- Taokang Institute of Neuro Medicine, Xiamen, Fujian, China
| | - Trevor Lee
- Department of Cell and Developmental Biology, Cornell University Weill Medical College, New York, NY, USA
| | - Yanxia Gao
- School of Life Sciences and Technology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhongyu Yuan
- School of Life Sciences and Technology, Shanghai Jiao Tong University, Shanghai, China
| | - Yanzhen Nie
- School of Life Sciences and Technology, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Sun
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| |
Collapse
|
37
|
Abstract
Mother's own milk provides personalized nutrition and immune protection to the developing infant. The presence of healthy microbes plays an important role in the infant's gut by programming the microbiota and excluding potential pathogens. This review describes the important components in mother's own milk that contribute to its superiority for infant nutrition and suggest potential strategies to replicate these factors in alternative feedings when sufficient milk is unavailable. Current strategies to supplement, substitute and replicate mother's own milk including microbial restoration, use of unpasteurized donor human milk, probiotics and fortification are discussed. Critical work remains to be done in understanding the human milk microbiome and metabolome and in improving lactation support for mothers of preterm infants. Increasing delivery of mother's own milk and milk components to infants would likely positively impact infant mortality and health worldwide.
Collapse
Affiliation(s)
- Evon DeBose-Scarlett
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, 2033 Mowry Road Rm 307, Gainesville, FL 32610 USA.
| | - Marion M Bendixen
- College of Nursing, University of Florida, PO Box 100197, Gainesville, FL 32610-0197 USA.
| | - Graciela L Lorca
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, 2033 Mowry Road Rm 307, Gainesville, FL 32610 USA.
| | - Leslie Ann Parker
- College of Nursing, University of Florida, PO Box 100197, Gainesville, FL 32610-0197 USA.
| |
Collapse
|
38
|
Amat S, Holman DB, Schmidt K, Menezes ACB, Baumgaertner F, Winders T, Kirsch JD, Liu T, Schwinghamer TD, Sedivec KK, Dahlen CR. The Nasopharyngeal, Ruminal, and Vaginal Microbiota and the Core Taxa Shared across These Microbiomes in Virgin Yearling Heifers Exposed to Divergent In Utero Nutrition during Their First Trimester of Gestation and in Pregnant Beef Heifers in Response to Mineral Supplementation. Microorganisms 2021; 9:2011. [PMID: 34683332 PMCID: PMC8537542 DOI: 10.3390/microorganisms9102011] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/11/2021] [Accepted: 09/20/2021] [Indexed: 12/26/2022] Open
Abstract
In the present study, we evaluated whether the nasopharyngeal, ruminal, and vaginal microbiota would diverge (1) in virgin yearling beef heifers (9 months old) due to the maternal restricted gain during the first trimester of gestation; and (2) in pregnant beef heifers in response to the vitamin and mineral (VTM) supplementation during the first 6 months of pregnancy. As a secondary objective, using the microbiota data obtained from these two cohorts of beef heifers managed at the same location and sampled at the same time, we performed a holistic assessment of the microbial ecology residing within the respiratory, gastrointestinal, and reproductive tract of cattle. Our 16S rRNA gene sequencing results revealed that both α and β-diversity of the nasopharyngeal, ruminal and vaginal microbiota did not differ between virgin heifers raised from dams exposed to either a low gain (targeted average daily gain of 0.28 kg/d, n = 22) or a moderate gain treatment (0.79 kg/d, n = 23) during the first 84 days of gestation. Only in the vaginal microbiota were there relatively abundant genera that were affected by maternal rate of gain during early gestation. Whilst there was no significant difference in community structure and diversity in any of the three microbiota between pregnant heifers received no VTM (n = 15) and VTM supplemented (n = 17) diets, the VTM supplementation resulted in subtle compositional alterations in the nasopharyngeal and ruminal microbiota. Although the nasopharyngeal, ruminal, and vaginal microbiota were clearly distinct, a total of 41 OTUs, including methanogenic archaea, were identified as core taxa shared across the respiratory, gastrointestinal, and reproductive tracts of both virgin and pregnant heifers.
Collapse
Affiliation(s)
- Samat Amat
- Department of Microbiological Sciences, North Dakota State University, Fargo, ND 58108, USA;
| | - Devin B. Holman
- Lacombe Research and Development Centre, Agriculture and Agri-Food Canada, 6000 C & E Trail, Lacombe, AB T4L 1W1, Canada; (D.B.H.); (T.L.)
| | - Kaycie Schmidt
- Department of Microbiological Sciences, North Dakota State University, Fargo, ND 58108, USA;
| | - Ana Clara B. Menezes
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58102, USA; (A.C.B.M.); (F.B.); (T.W.); (J.D.K.); (C.R.D.)
| | - Friederike Baumgaertner
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58102, USA; (A.C.B.M.); (F.B.); (T.W.); (J.D.K.); (C.R.D.)
| | - Thomas Winders
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58102, USA; (A.C.B.M.); (F.B.); (T.W.); (J.D.K.); (C.R.D.)
| | - James D. Kirsch
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58102, USA; (A.C.B.M.); (F.B.); (T.W.); (J.D.K.); (C.R.D.)
| | - Tingting Liu
- Lacombe Research and Development Centre, Agriculture and Agri-Food Canada, 6000 C & E Trail, Lacombe, AB T4L 1W1, Canada; (D.B.H.); (T.L.)
| | - Timothy D. Schwinghamer
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB T1J 4B1, Canada;
| | - Kevin K. Sedivec
- Central Grasslands Research Extension Center, North Dakota State University, Streeter, ND 58483, USA;
| | - Carl R. Dahlen
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58102, USA; (A.C.B.M.); (F.B.); (T.W.); (J.D.K.); (C.R.D.)
| |
Collapse
|
39
|
Ta LDH, Tay CJX, Lay C, de Sessions PF, Tan CPT, Tay MJY, Lau HX, Zulkifli AB, Yap GC, Tham EH, Ho EXP, Goh AEN, Godfrey KM, Eriksson JG, Knol J, Gluckman PD, Chong YS, Chan JKY, Tan KH, Chong KW, Goh SH, Cheng ZR, Lee BW, Shek LPC, Loo EXL. Household environmental microbiota influences early-life eczema development. Environ Microbiol 2021; 23:7710-7722. [PMID: 34309161 DOI: 10.1111/1462-2920.15684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/20/2021] [Accepted: 07/19/2021] [Indexed: 11/28/2022]
Abstract
Exposure to a diverse microbial environment during pregnancy and early postnatal period is important in determining predisposition towards allergy. However, the effect of environmental microbiota exposure during preconception, pregnancy and postnatal life on development of allergy in the child has not been investigated so far. In the S-PRESTO (Singapore PREconception Study of long Term maternal and child Outcomes) cohort, we collected house dust during all three critical window periods and analysed microbial composition using 16S rRNA gene sequencing. At 6 and 18 months, the child was assessed for eczema by clinicians. In the eczema group, household environmental microbiota was characterized by presence of human-associated bacteria Actinomyces, Anaerococcus, Finegoldia, Micrococcus, Prevotella and Propionibacterium at all time points, suggesting their possible contributions to regulating host immunity and increasing the susceptibility to eczema. In the home environment of the control group, putative protective effect of an environmental microbe Planomicrobium (Planococcaceae family) was observed to be significantly higher than that in the eczema group. Network correlation analysis demonstrated inverse relationships between beneficial Planomicrobium and human-associated bacteria (Actinomyces, Anaerococcus, Finegoldia, Micrococcus, Prevotella and Propionibacterium). Exposure to natural environmental microbiota may be beneficial to modulate shed human-associated microbiota in an indoor environment.
Collapse
Affiliation(s)
- Le Duc Huy Ta
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Carina Jing Xuan Tay
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christophe Lay
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Danone Nutricia Research, Singapore, Singapore
| | - Paola Florez de Sessions
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Cheryl Pei Ting Tan
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Michelle Jia Yu Tay
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hui Xing Lau
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Atiqa Binte Zulkifli
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Gaik Chin Yap
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Elizabeth Huiwen Tham
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
| | - Eliza Xin Pei Ho
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Anne Eng Neo Goh
- Allergy Service, Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Keith M Godfrey
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, UK.,Medical Research Council Life course Epidemiology Unit, Southampton, SO16 6YD, UK
| | - Johan G Eriksson
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.,Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore.,Folkhälsan Research Center, Helsinki, Finland.,Department of General Practice and Primary Health Care, University of Helsinki, Finland
| | - Jan Knol
- Danone Nutricia Research, Utrecht, The Netherlands.,Wageningen University, Wageningen, The Netherlands
| | - Peter D Gluckman
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.,Liggins Institute, University of Auckland, New Zealand
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.,Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Kok Hian Tan
- Department of Maternal Fetal Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Kok Wee Chong
- Allergy Service, Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Si Hui Goh
- Allergy Service, Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Zai Ru Cheng
- Respiratory Medicine Service, Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Bee Wah Lee
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lynette Pei-Chi Shek
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
| | - Evelyn Xiu Ling Loo
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
40
|
Li JH, Liu JL, Zhang KK, Chen LJ, Xu JT, Xie XL. The Adverse Effects of Prenatal METH Exposure on the Offspring: A Review. Front Pharmacol 2021; 12:715176. [PMID: 34335277 PMCID: PMC8317262 DOI: 10.3389/fphar.2021.715176] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/01/2021] [Indexed: 01/12/2023] Open
Abstract
Abuse of methamphetamine (METH), an illicit psychostimulant, is a growing public health issue. METH abuse during pregnancy is on the rise due to its stimulant, anorectic, and hallucinogenic properties. METH can lead to multiple organ toxicity in adults, including neurotoxicity, cardiovascular toxicity, and hepatotoxicity. It can also cross the placental barrier and have long-lasting effects on the fetus. This review summarizes neurotoxicity, cardiovascular toxicity, hepatotoxicity, toxicity in other organs, and biomonitoring of prenatal METH exposure, as well as the possible emergence of sensitization associated with METH. We proposed the importance of gut microbiota in studying prenatal METH exposure. There is rising evidence of the adverse effects of METH exposure during pregnancy, which are of significant concern.
Collapse
Affiliation(s)
- Jia-Hao Li
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Jia-Li Liu
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Kai-Kai Zhang
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Li-Jian Chen
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Jing-Tao Xu
- Department of Forensic Clinical Medicine, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Xiao-Li Xie
- Department of Toxicology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, Guangzhou, China
| |
Collapse
|
41
|
Tal S, Tikhonov E, Aroch I, Hefetz L, Turjeman S, Koren O, Kuzi S. Developmental intestinal microbiome alterations in canine fading puppy syndrome: a prospective observational study. NPJ Biofilms Microbiomes 2021; 7:52. [PMID: 34162880 PMCID: PMC8222291 DOI: 10.1038/s41522-021-00222-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 05/20/2021] [Indexed: 01/01/2023] Open
Abstract
Fading puppy syndrome (FPS) is a fatal condition in neonatal dogs. Intestinal microbial alterations, although never investigated, may be involved in its pathophysiology. The study examined the occurrence of FPS and its associations with dam, puppy, and husbandry characteristics, compared the intestinal microbial diversity of healthy puppies and those with FPS, and examined whether intestinal microbiomes are predictive of FPS. Day 1 and 8 post-partum (PP) rectal swabs were collected from healthy puppies and puppies which later developed FPS. Microbial compositional structure, including alpha and beta diversities and relative abundance of specific taxa were compared between groups, and microbial data was applied to a machine-learning model to assess the predictive performance of microbial indices of FPS or death. FPS occurred in 22/165 puppies (13%), with a 100% mortality rate. FPS was associated (P < 0.001) with decreased Day 1 PP puppy activity. Day 1 (P = 0.003) and 8 (P = 0.005) PP rectal beta diversities were different in puppies with FPS vs healthy ones. Increased Proteobacteria/Firmicutes ratio, increased relative abundance of Pasteurellaceae, and decreased relative abundance of Clostridia and Enterococcus were associated with FPS. A machine-learning model showed that Day 1 PP rectal microbiome composition accurately predicted FPS-related death. We found that specific rectal microbial phenotypes are associated with FPS, reflecting the significant role of microbiome alterations in this phenomenon. These findings may serve as useful microbial indices for early diagnosis of puppies at risk of FPS and may provide specific therapeutic targets.
Collapse
Affiliation(s)
- Smadar Tal
- The Hebrew University Veterinary Teaching Hospital and Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | | | - Itamar Aroch
- The Hebrew University Veterinary Teaching Hospital and Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Lior Hefetz
- The Hebrew University Veterinary Teaching Hospital and Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Sondra Turjeman
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Sharon Kuzi
- The Hebrew University Veterinary Teaching Hospital and Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel.
| |
Collapse
|
42
|
Bardanzellu F, Puddu M, Peroni DG, Fanos V. The clinical impact of maternal weight on offspring health: lights and shadows in breast milk metabolome. Expert Rev Proteomics 2021; 18:571-606. [PMID: 34107825 DOI: 10.1080/14789450.2021.1940143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Pre-pregnancy overweight and obesity, depending on maternal nutrition and metabolic state, can influence fetal, neonatal and long-term offspring health, regarding cardio-metabolic, respiratory, immunological and cognitive outcomes. Thus, maternal weight can act, through mechanisms that are not full understood, on the physiology and metabolism of some fetal organs and tissues, to adapt themselves to the intrauterine environment and nutritional reserves. These effects could occur by modulating gene expression, neonatal microbiome, and through breastfeeding. AREAS COVERED In this paper, we investigated the potential effects of metabolites found altered in breast milk (BM) of overweight/obese mothers, through an extensive review of metabolomics studies, and the potential short- and long-term clinical effects in the offspring, especially regarding overweight, glucose homeostasis, insulin resistance, oxidative stress, infections, immune processes, and neurodevelopment. EXPERT OPINION Metabolomics seems the ideal tool to investigate BM variation depending on maternal or fetal/neonatal factors. In particular, BM metabolome alterations according to maternal conditions were recently pointed out in cases of gestational diabetes, preeclampsia, intrauterine growth restriction and maternal overweight/obesity. In our opinion, even if BM is the food of choice in neonatal nutrition, the deepest comprehension of its composition in overweight/obese mothers could allow targeted supplementation, to improve offspring health and metabolic homeostasis.
Collapse
Affiliation(s)
- Flaminia Bardanzellu
- Neonatal Intensive Care Unit, Department of Surgical Sciences, AOU and University of Cagliari. SS 554 km 4,500, 09042 Monserrato. Italy
| | - Melania Puddu
- Neonatal Intensive Care Unit, Department of Surgical Sciences, AOU and University of Cagliari. SS 554 km 4,500, 09042 Monserrato. Italy
| | - Diego Giampietro Peroni
- Clinical and Experimental Medicine Department, section of Pediatrics, University of Pisa, Italy. Via Roma, 55, 56126 Pisa PI, Italy
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Department of Surgical Sciences, AOU and University of Cagliari. SS 554 km 4,500, 09042 Monserrato. Italy
| |
Collapse
|
43
|
He Q, Zou T, Chen J, He J, Jian L, Xie F, You J, Wang Z. Methyl-Donor Micronutrient for Gestating Sows: Effects on Gut Microbiota and Metabolome in Offspring Piglets. Front Nutr 2021; 8:675640. [PMID: 34164424 PMCID: PMC8215270 DOI: 10.3389/fnut.2021.675640] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/22/2021] [Indexed: 12/17/2022] Open
Abstract
This study aimed to investigate the effects of maternal methyl-donor micronutrient supplementation during gestation on gut microbiota and the fecal metabolic profile in offspring piglets. Forty-three Duroc × Erhualian gilts were assigned to two dietary groups during gestation: control diet (CON) and CON diet supplemented with MET (folic acid, methionine, choline, vitamin B6, and vitamin B12). The body weights of offspring piglets were recorded at birth and weaning. Besides this, fresh fecal samples of offspring piglets were collected at 7, 14, and 21 days. The gut microbiota composition, metabolic profile, and short-chain fatty acid (SCFA) profiles in the fecal samples were determined using 16S rDNA sequencing, liquid chromatography-mass spectrometry metabolomics, and gas chromatography methods, respectively. The results showed that maternal methyl-donor micronutrient supplementation increased the microbiota diversity and uniformity in feces of offspring piglets as indicated by increased Shannon and Simpson indices at 7 days, and greater Simpson, ACE, Chao1 and observed species indices at 21 days. Specifically, at the phylum level, the relative abundance of Firmicutes and the Firmicutes to Bacteroidetes ratio were elevated by maternal treatment. At the genus level, the relative abundance of SCFA-producing Dialister, Megasphaera, and Turicibacter, and lactate-producing Sharpea as well as Akkermansia, Weissella, and Pediococcus were increased in the MET group. The metabolic analyses show that maternal methyl-donor micronutrient addition increased the concentrations of individual and total SCFAs of 21-day piglets and increased metabolism mainly involving amino acids, pyrimidine, and purine biosynthesis. Collectively, maternal methyl-donor micronutrient addition altered gut microbiota and the fecal metabolic profile, resulting in an improved weaning weight of offspring piglets.
Collapse
Affiliation(s)
- Qin He
- Key Laboratory of Animal Nutrition in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China.,Key Innovation Center for Industry-Education Integration of High-Quality and Safety Livestock Production in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China
| | - Tiande Zou
- Key Laboratory of Animal Nutrition in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China.,Key Innovation Center for Industry-Education Integration of High-Quality and Safety Livestock Production in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China
| | - Jun Chen
- Key Laboratory of Animal Nutrition in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China.,Key Innovation Center for Industry-Education Integration of High-Quality and Safety Livestock Production in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China
| | - Jia He
- Key Laboratory of Animal Nutrition in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China.,Key Innovation Center for Industry-Education Integration of High-Quality and Safety Livestock Production in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China
| | - Li Jian
- Key Laboratory of Animal Nutrition in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China.,Key Innovation Center for Industry-Education Integration of High-Quality and Safety Livestock Production in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China
| | - Fei Xie
- Key Laboratory of Animal Nutrition in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China.,Key Innovation Center for Industry-Education Integration of High-Quality and Safety Livestock Production in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China
| | - Jinming You
- Key Laboratory of Animal Nutrition in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China.,Key Innovation Center for Industry-Education Integration of High-Quality and Safety Livestock Production in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China
| | - Zirui Wang
- Key Laboratory of Animal Nutrition in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China.,Key Innovation Center for Industry-Education Integration of High-Quality and Safety Livestock Production in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China
| |
Collapse
|
44
|
Chen CM, Chou HC, Yang YCSH. Maternal Antibiotic Treatment Disrupts the Intestinal Microbiota and Intestinal Development in Neonatal Mice. Front Microbiol 2021; 12:684233. [PMID: 34177871 PMCID: PMC8220820 DOI: 10.3389/fmicb.2021.684233] [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: 03/23/2021] [Accepted: 05/04/2021] [Indexed: 12/20/2022] Open
Abstract
Maternal antibiotic treatment (MAT) during prenatal and intrapartum periods alters the bacterial composition and diversity of the intestinal microbiota of the offspring. The effect of MAT during pregnancy on the intestinal microbiota and its relationship with intestinal development remain unknown. This study investigated the effects of MAT during pregnancy on intestinal microbiota, injury and inflammation, vascularization, cellular proliferation, and the intestinal barrier in neonatal mice. At timed intervals, we fed pregnant C57BL/6N mice sterile drinking water containing antibiotics (ampicillin, gentamicin, and vancomycin; all 1 mg/ml) from gestational day 15 to delivery. The control dams were fed sterile drinking water. Antibiotic administration was halted immediately after birth. On postnatal day 7, the intestinal microbiota was sampled from the lower gastrointestinal tract and the ileum was harvested for histology, Western blot, and cytokines analyses. MAT significantly reduced the relative abundance of Bacteroidetes and Firmicutes and significantly increased the relative abundance of Proteobacteria in the intestine compared with their abundances in the control group. MAT also significantly increased intestinal injury score and cytokine levels, reduced the number of intestinal goblet cells and proliferating cell nuclear antigen-positive cells, and reduced the expressions of vascular endothelial growth factor and tight junction proteins. Therefore, we proposed that maternal antibiotic exposure during pregnancy disrupts the intestinal microbiota and intestinal development in neonatal mice.
Collapse
Affiliation(s)
- Chung-Ming Chen
- Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiu-Chu Chou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chen S H Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
45
|
Diet-induced dysbiosis of the maternal gut microbiome in early life programming of neurodevelopmental disorders. Neurosci Res 2021; 168:3-19. [PMID: 33992660 DOI: 10.1016/j.neures.2021.05.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/10/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022]
Abstract
The maternal gut microbiome plays a critical role in fetal and early postnatal development, shaping fundamental processes including immune maturation and brain development, among others. Consequently, it also contributes to fetal programming of health and disease. Over the last decade, epidemiological studies and work in preclinical animal models have begun to uncover a link between dysbiosis of the maternal gut microbiome and neurodevelopmental disorders in offspring. Neurodevelopmental disorders are caused by both genetic and environmental factors, and their interactions; however, clinical heterogeneity, phenotypic variability, and comorbidities make identification of underlying mechanisms difficult. Among environmental factors, exposure to maternal obesity in utero confers a significant increase in risk for neurodevelopmental disorders. Obesogenic diets in humans, non-human primates, and rodents induce functional modifications in maternal gut microbiome composition, which animal studies suggest are causally related to adverse mental health outcomes in offspring. Here, we review evidence linking maternal diet-induced gut dysbiosis to neurodevelopmental disorders and discuss how it could affect pre- and early postnatal brain development. We are hopeful that this burgeoning field of research will revolutionize antenatal care by leading to accessible prophylactic strategies, such as prenatal probiotics, to improve mental health outcomes in children affected by maternal diet-induced obesity.
Collapse
|
46
|
Littlejohn P, Finlay BB. When a pandemic and an epidemic collide: COVID-19, gut microbiota, and the double burden of malnutrition. BMC Med 2021; 19:31. [PMID: 33504332 PMCID: PMC7840385 DOI: 10.1186/s12916-021-01910-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/13/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND It is estimated that the COVID-19 pandemic will drastically increase all forms of malnutrition. Of particular concern, yet understated, is the potential to increase the double burden of malnutrition (DBM) epidemic. This coexistence of undernutrition together with overweight and obesity, or diet-related non-communicable disease (NCD), within low- to middle-income countries (LMICs) is increasing rapidly. Although multiple factors contribute to the DBM, food insecurity (FI) and gut microbiota dysbiosis play a crucial role. Both under- and overnutrition have been shown to be a consequence of food insecurity. The gut microbiota has also been recently implicated in playing a role in under- and overnutrition, with altered community structure and function common to both. The pandemic has already caused significant shifts in food availability which has immediate effects on the gut microbiome. In this opinion paper, we discuss how COVID-19 may indirectly exacerbate the DBM through food insecurity and the gut microbiome. MAIN TEXT The World Food Programme (WFP) estimates that 265 million people in LMICs will experience acute hunger in 2020 due to the pandemic, nearly doubling the original projection of 135 million. Global border closures to food trade, loss of food production, and stark decline in household income will exacerbate starvation while simultaneously necessitating that families resort to calorie-dense, nutrient-poor foods, thereby increasing obesity. While food insecurity, which is the persistent lack of consistent access to adequate and nutrient-rich foods, will primarily drive nutrition behavior, the gut microbiome is perhaps a key biological mechanism. Numerous human and animal studies describe low diversity and an increase in inflammatory species as characteristic features of the undernourished and overnourished gut microbiota. Indeed, fecal transplant studies show that microbiota transfer from undernourished and overnourished humans to germ-free mice lacking a microbiome transfers the physical and metabolic phenotype, suggesting a causal role for the microbiota in under- and overnutrition. The observed microbiome dysbiosis within severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) coupled with the DBM presents a viscous cycle. CONCLUSION Low- to mid-income countries will likely see an increase in the DBM epidemic. Providing access to nutritious foods and protecting individuals' gut microbiome to "flatten the curve" of the DBM trajectory should be prioritized.
Collapse
Affiliation(s)
- Paula Littlejohn
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, V6T 1Z3, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, V6T 1Z4, Canada
| | - B Brett Finlay
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, V6T 1Z3, Canada.
- Michael Smith Laboratories, University of British Columbia, Vancouver, V6T 1Z4, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, V6T 1Z3, Canada.
| |
Collapse
|
47
|
Mechanisms Underlying the Cognitive and Behavioural Effects of Maternal Obesity. Nutrients 2021; 13:nu13010240. [PMID: 33467657 PMCID: PMC7829712 DOI: 10.3390/nu13010240] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/11/2021] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
The widespread consumption of 'western'-style diets along with sedentary lifestyles has led to a global epidemic of obesity. Epidemiological, clinical and preclinical evidence suggests that maternal obesity, overnutrition and unhealthy dietary patterns programs have lasting adverse effects on the physical and mental health of offspring. We review currently available preclinical and clinical evidence and summarise possible underlying neurobiological mechanisms by which maternal overnutrition may perturb offspring cognitive function, affective state and psychosocial behaviour, with a focus on (1) neuroinflammation; (2) disrupted neuronal circuities and connectivity; and (3) dysregulated brain hormones. We briefly summarise research implicating the gut microbiota in maternal obesity-induced changes to offspring behaviour. In animal models, maternal obesogenic diet consumption disrupts CNS homeostasis in offspring, which is critical for healthy neurodevelopment, by altering hypothalamic and hippocampal development and recruitment of glial cells, which subsequently dysregulates dopaminergic and serotonergic systems. The adverse effects of maternal obesogenic diets are also conferred through changes to hormones including leptin, insulin and oxytocin which interact with these brain regions and neuronal circuits. Furthermore, accumulating evidence suggests that the gut microbiome may directly and indirectly contribute to these maternal diet effects in both human and animal studies. As the specific pathways shaping abnormal behaviour in offspring in the context of maternal obesogenic diet exposure remain unknown, further investigations are needed to address this knowledge gap. Use of animal models permits investigation of changes in neuroinflammation, neurotransmitter activity and hormones across global brain network and sex differences, which could be directly and indirectly modulated by the gut microbiome.
Collapse
|
48
|
Christoforou ER, Sferruzzi-Perri AN. Molecular mechanisms governing offspring metabolic programming in rodent models of in utero stress. Cell Mol Life Sci 2020; 77:4861-4898. [PMID: 32494846 PMCID: PMC7658077 DOI: 10.1007/s00018-020-03566-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/23/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022]
Abstract
The results of different human epidemiological datasets provided the impetus to introduce the now commonly accepted theory coined as 'developmental programming', whereby the presence of a stressor during gestation predisposes the growing fetus to develop diseases, such as metabolic dysfunction in later postnatal life. However, in a clinical setting, human lifespan and inaccessibility to tissue for analysis are major limitations to study the molecular mechanisms governing developmental programming. Subsequently, studies using animal models have proved indispensable to the identification of key molecular pathways and epigenetic mechanisms that are dysregulated in metabolic organs of the fetus and adult programmed due to an adverse gestational environment. Rodents such as mice and rats are the most used experimental animals in the study of developmental programming. This review summarises the molecular pathways and epigenetic mechanisms influencing alterations in metabolic tissues of rodent offspring exposed to in utero stress and subsequently programmed for metabolic dysfunction. By comparing molecular mechanisms in a variety of rodent models of in utero stress, we hope to summarise common themes and pathways governing later metabolic dysfunction in the offspring whilst identifying reasons for incongruencies between models so to inform future work. With the continued use and refinement of such models of developmental programming, the scientific community may gain the knowledge required for the targeted treatment of metabolic diseases that have intrauterine origins.
Collapse
Affiliation(s)
- Efthimia R Christoforou
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Downing Site, Cambridge, UK
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Downing Site, Cambridge, UK.
| |
Collapse
|
49
|
Byrne DF, Geraghty AA, Yelverton CA, Murphy EF, Van Sinderen D, Cotter PD, McAuliffe FM. The impact of probiotic supplementation on metabolic health in healthy women of reproductive age: a systematic review. Food Funct 2020; 11:10279-10289. [PMID: 33174573 DOI: 10.1039/d0fo01727b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Products containing probiotics are targeted at healthy or at-risk individuals as a preventative measure to minimise disease risk. Most studies assessing the efficacy of probiotics in humans include a mixture of healthy and unhealthy populations, while studies that focus solely on female populations are largely limited to pregnancy or those with health conditions. Pre-conception is a significant time-point during the life-course, and improving female health status during this period may positively influence future offspring. The objective of this review is to assess the effect of probiotics administered in oral capsule formulation, on metabolic and immune markers in healthy, non-pregnant women of reproductive age. This review followed the PRISMA guidelines. Pubmed, EMBASE, CINAHL, and Web of Science were searched for relevant studies. English language articles relating to randomised-controlled trials were included. The search returned 3250 publications after duplicates were removed. Title (2516), abstract (642), and full text (87) screening excluded 3993 studies from consideration. Five papers were identified with outcomes of interest, and analysis of these showed no conclusive evidence that probiotic capsule supplementation elicited positive effects in this healthy population. This study highlights the need for further research to investigate the role that probiotics play during the pre-conception period, on female metabolic and immune health.
Collapse
Affiliation(s)
- David F Byrne
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Ireland.
| | | | | | | | | | | | | |
Collapse
|
50
|
Pérez-Cano FJ. What Does Influence the Neonatal Microbiome? Nutrients 2020; 12:nu12082472. [PMID: 32824433 PMCID: PMC7468762 DOI: 10.3390/nu12082472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 08/11/2020] [Indexed: 12/25/2022] Open
Abstract
This editorial aims to provide a concise summary of the factors involved in the dynamics of microbiome establishment and maturation. At the same time, it briefly updates the current knowledge and opens new questions in this regard. Many factors act as drivers of the microbiota’s development at both pre- and post-natal levels (e.g., maternal factors, antibiotic usage, type of delivery, dietary pattern, post-natal feeding type, etc.). However, it is interesting to research into its real impact, the relationship between these external modulators, and how to modulate them. The are great opportunities for new research in the field.
Collapse
Affiliation(s)
- Francisco J. Pérez-Cano
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; ; Tel.: +349-340-24505
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|