1
|
Redgwell TA, Thorsen J, Petit MA, Deng L, Vestergaard G, Russel J, Chawes B, Bønnelykke K, Bisgaard H, Nielsen DS, Sørensen S, Stokholm J, Shah SA. Prophages in the infant gut are pervasively induced and may modulate the functionality of their hosts. NPJ Biofilms Microbiomes 2025; 11:46. [PMID: 40108202 PMCID: PMC11923282 DOI: 10.1038/s41522-025-00674-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025] Open
Abstract
Gut microbiome (GM) composition and function is pivotal for human health and disease, of which the virome's importance is increasingly recognised. However, prophages and their induction patterns in the infant gut remain understudied. Here, we identified 10645 putative prophages in 662 metagenomes from 1-year-old children in the COPSAC2010 mother-child cohort and investigated their potential functions. No core provirome was found as the most prevalent vOTU was identified in only ~70% of the samples. The most dominant cluster of vOTUs in the cohort was related to Bacteroides phage Hanky p00', and it carried both diversity generating retroelements and genes involved in capsular polysaccharide synthesis. Paired analysis of viromes and metagenomes from the same samples revealed that most prophages within the infant gut were induced and that induction was unaffected by a range of environmental perturbers. In summary, prophages are major components of the infant gut that may have far reaching influences on the microbiome and its host.
Collapse
Affiliation(s)
- Tamsin A Redgwell
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Ledreborg Allé 34, DK-2820, Gentofte, Denmark
| | - Jonathan Thorsen
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Ledreborg Allé 34, DK-2820, Gentofte, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marie-Agnès Petit
- Micalis institute, INRAE, Agroparistech, Université Paris-Saclay, Jouy en Josas, France
| | - Ling Deng
- Section of Food Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26, 1958, Frederiksberg C, Denmark
| | - Gisle Vestergaard
- Technical University of Denmark, Section of Bioinformatics, Department of Health Technology, 2800 Kgs, Lyngby, Denmark
| | - Jakob Russel
- Department of Biology, Section of Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Bo Chawes
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Ledreborg Allé 34, DK-2820, Gentofte, Denmark
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Ledreborg Allé 34, DK-2820, Gentofte, Denmark
| | - Hans Bisgaard
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Ledreborg Allé 34, DK-2820, Gentofte, Denmark
| | - Dennis S Nielsen
- Section of Food Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26, 1958, Frederiksberg C, Denmark
| | - Søren Sørensen
- Department of Biology, Section of Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Ledreborg Allé 34, DK-2820, Gentofte, Denmark
- Section of Food Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26, 1958, Frederiksberg C, Denmark
| | - Shiraz A Shah
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Ledreborg Allé 34, DK-2820, Gentofte, Denmark.
| |
Collapse
|
2
|
Shi Y, Yin R, Pang J, Chen Y, Li Z, Su S, Wen Y. Impact of complementary feeding on infant gut microbiome, metabolites and early development. Food Funct 2024; 15:10663-10678. [PMID: 39354871 DOI: 10.1039/d4fo03948c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Introducing complementary foods is critical for promoting infant health and development. During the weaning period, the dietary patterns provide essential nutrients and facilitate the development of a diverse gut microbiome, which plays significant roles in the regulation of immune, metabolic, and neurological functions. This study enrolled 200 families to assess the impact of complementary feeding on infant growth and health outcomes. Data included detailed records of feeding practices, infant growth measurements, health assessments, and fecal samples and breast milk collected between weeks 12 and 32 postpartum. The gut microbiome was analyzed using 16S rRNA sequencing, while metabolites such as human milk oligosaccharides (HMOs), monosaccharides, and short-chain fatty acids (SCFAs) were measured using chromatography-mass spectrometry. Results revealed a high prevalence of breastfeeding, with complementary food introduced at around 16 weeks. Significant alterations in the infant gut microbiome were observed, particularly in the genera Lactobacillus, Akkermansia, and Staphylococcus. Additionally, the levels of HMOs, monosaccharides, and SCFAs were found to be influenced by the introduction of complementary foods. Significant correlations emerged between complementary feeding practices, gut microbiome diversity, specific bacterial genera (e.g., Streptococcus, Lactobacillus, Bifidobacterium, and Clostridioides), and key metabolites (such as lacto-N-tetraose, lacto-N-neotetraose, mannose, and butyric acid). This study offers valuable insights into the complex interactions between complementary feeding, gut microbiome development, and metabolite profiles during early infant growth. Future research with larger cohorts and targeted dietary interventions is recommended to further elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Yudong Shi
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Ran Yin
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Jinzhu Pang
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Yun Chen
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Zhouyong Li
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Shengpeng Su
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Yongping Wen
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| |
Collapse
|
3
|
Majumder A, Bano S. How the Western Diet Thwarts the Epigenetic Efforts of Gut Microbes in Ulcerative Colitis and Its Association with Colorectal Cancer. Biomolecules 2024; 14:633. [PMID: 38927037 PMCID: PMC11201633 DOI: 10.3390/biom14060633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Ulcerative colitis (UC) is an autoimmune disease in which the immune system attacks the colon, leading to ulcer development, loss of colon function, and bloody diarrhea. The human gut ecosystem consists of almost 2000 different species of bacteria, forming a bioreactor fueled by dietary micronutrients to produce bioreactive compounds, which are absorbed by our body and signal to distant organs. Studies have shown that the Western diet, with fewer short-chain fatty acids (SCFAs), can alter the gut microbiome composition and cause the host's epigenetic reprogramming. Additionally, overproduction of H2S from the gut microbiome due to changes in diet patterns can further activate pro-inflammatory signaling pathways in UC. This review discusses how the Western diet affects the microbiome's function and alters the host's physiological homeostasis and susceptibility to UC. This article also covers the epidemiology, prognosis, pathophysiology, and current treatment strategies for UC, and how they are linked to colorectal cancer.
Collapse
Affiliation(s)
- Avisek Majumder
- Department of Medicine, University of California, San Francisco, CA 94158, USA
| | | |
Collapse
|
4
|
Ajibola O, Penumutchu S, Gulumbe B, Aminu U, Belenky P. Longitudinal Analysis of the Impacts of Urogenital Schistosomiasis on the Gut microbiota of Adolescents in Nigeria. RESEARCH SQUARE 2023:rs.3.rs-2832346. [PMID: 37163079 PMCID: PMC10168446 DOI: 10.21203/rs.3.rs-2832346/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The gut microbiome is important for many host physiological processes and helminths and these interactions may lead to microbial changes. We carried out a longitudinal study of the impacts of S. haematobium infection on the gut microbiome of adolescents (11-15 years) in northern Nigeria pre and post praziquantel treatment. Using 16S sequencing a total of 267 DNA from faecal samples of infected versus uninfected adolescents were amplified and sequenced on an Illumina Miseq. We assessed the diversity of the taxa using alpha diversity metrices and observed that using Shannon index we obtained significant differences when we compared infected samples at 3, 9 and 12 months to baseline uninfected controls (P= <0.0001, P=0.0342 and P=0.0003 respectively). Microbial community composition analysis revealed that there were only significant differences at 3, 9 and 12 months (P=0.001, P=0.001, P=0.001 and P=0.001, respectively). We also demonstrated that the effects of the infection on the gut was more significant than praziquantel. Overall, our data suggests that S. haematobium, a non-gut resident parasite has indirect interactions with the gut. The bacterial taxa changes we have identified opens up the opportunity to investigate their role in human health, especially in urogenital schistosomiasis endemic communities.
Collapse
|
5
|
Yin N, Liu X, Zhang X, Wen J, Ma H, Yin X, Xie C, Hou Y, Wang J. Comparison of the effects of different infant formulas on the growth and development and intestinal flora of infants. Food Sci Nutr 2023; 11:1113-1126. [PMID: 36789049 PMCID: PMC9922136 DOI: 10.1002/fsn3.3149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 11/05/2022] [Accepted: 11/06/2022] [Indexed: 12/12/2022] Open
Abstract
The purpose of this study was to compare the effects of different infant formulas on the growth and development, sleep, allergy symptoms, and intestinal flora of infants. A total of 428 infants participated in the study. Breastfeeding (BF) was used as the control, and the remaining subjects were randomly assigned to the full goat milk protein formula group (FGM), partial goat milk protein formula group (PGM), and cow milk formula group (M). During the 6-month feeding experiment, data on the growth, sleep, allergy symptoms, and intestinal flora of infants were collected using questionnaires, anthropometric measurements, and biochemical examinations. In general, the basic information of the participants was consistent among the groups. There were no differences in infant weight, length, or head circumference among the groups (p > .05). The sleep time of infants in the formula-fed groups was longer than that of the breastfeeding group at baseline (p < .05), but there were no differences at mid-term or outcome (p > .05). The incidence of allergic symptoms continued to decrease, and the total scores of allergic symptoms did not differ among the groups (p > .05). The relative abundance of intestinal Bifidobacteriaceae in the PGM group was lower than that in the other groups (p < .05). There was no difference in the β-diversity of intestinal flora between formula-fed and breastfed infants (p > .05). There were strong correlations in the composition of the main intestinal flora at the family level between the formula and breastfeeding groups. This study showed that within 6 months of feeding, there were no significant differences in the growth and development, allergic symptoms, or intestinal flora of the infants among the groups.
Collapse
Affiliation(s)
- Ning Yin
- Department of Nutrition and Food Hygiene, School of Public HealthPeking UniversityBeijingChina
| | - Xinran Liu
- Department of Nutrition and Food Hygiene, School of Public HealthPeking UniversityBeijingChina
| | - Xiaoxuan Zhang
- Department of Nutrition and Food Hygiene, School of Public HealthPeking UniversityBeijingChina
| | - Jing Wen
- Department of Nutrition and Food Hygiene, School of Public HealthPeking UniversityBeijingChina
| | - Huijuan Ma
- Department of Nutrition and Food Hygiene, School of Public HealthPeking UniversityBeijingChina
| | - Xueqian Yin
- Department of Nutrition and Food Hygiene, School of Public HealthPeking UniversityBeijingChina
| | | | - Yanmei Hou
- Ausnutria Hyproca Nutrition Co., Ltd.ChangshaChina
| | - Junbo Wang
- Department of Nutrition and Food Hygiene, School of Public HealthPeking UniversityBeijingChina
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food SafetyPeking UniversityBeijingChina
| |
Collapse
|
6
|
Wang Y, Li X, Chen X, Kulyar MFEA, Duan K, Li H, Bhutta ZA, Wu Y, Li K. Gut Fungal Microbiome Responses to Natural Cryptosporidium Infection in Horses. Front Microbiol 2022; 13:877280. [PMID: 35875530 PMCID: PMC9298756 DOI: 10.3389/fmicb.2022.877280] [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: 02/22/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
It is critical to characterize changes in the structure and composition of the host fungal community in natural Cryptosporidium infection, because it gives the possible overview of gut microbiome in host homeostasis and disease progression. A total of 168 rectal fecal samples were collected and examined using nPCR. The positive samples were double-checked using 18S rDNA high-throughput sequencing. After confirmation, ITS high-throughput sequencing was utilized to investigate the fungal community’s response to natural Cryptosporidium infection. Results showed that a total of three positive samples (1.79%) were identified with an increased abundance of fungi associated with health hazards, such as class Dothideomycetes, families, i.e., Cladosporiaceae, Glomerellaceae, and genera, i.e., Wickerhamomyces, Talaromyces, Cladosporium, Dactylonectria, and Colletotrichum. On the contrary, taxa associated with favorable physiological effects on the host were shown to have the reverse impact, such as families, i.e., Psathyrellaceae, Pseudeurotiaceae and genera (Beauveria, Nigrospora, and Diversispora). For the first time, we evaluated the condition of natural Cryptosporidium infection in horses in Wuhan, China, and discovered distinct variations in the fungal microbiome in response to natural infection. It might prompt a therapy or prevention strategy to apply specific fungal microorganisms that are probably responsible for decreased susceptibility or increased resistance to infection.
Collapse
Affiliation(s)
- Yaping Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xuwen Li
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiushuang Chen
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | | | - Kun Duan
- China Tobacco Henan Industrial Co., Ltd., Zhengzhou, China
| | - Huade Li
- Sichuan Academy of Grassland Science, Chengdu, China
| | - Zeeshan Ahmad Bhutta
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, South Korea
| | - Yi Wu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Kun Li
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
7
|
Cui Z, Li J, Zhen Y, Fan P, Du G. The Effect of Whole-Grain Diet on the Gut Microbiota of the Elderly Individuals. Front Nutr 2022; 9:919838. [PMID: 35832054 PMCID: PMC9273149 DOI: 10.3389/fnut.2022.919838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/31/2022] [Indexed: 11/20/2022] Open
Abstract
A whole-grain (WG) diet affects human health in multiple ways. However, the effect of WG on the gut microbiota of the elderly individuals is still largely unknown. In this study, WG did not affect the microbial α-diversity but had a profound impact on the microbes' abundance in the elderly individuals. WG increased the abundance of Verrucomicrobia and decreased the abundance of Firmicutes. The prediction of microbial function showed that glucose metabolism and lipid metabolism were inhibited. In addition, the effects of WG on the gut microbiota of normal-weight (NW) and overweight (OW) individuals were different. WG increased Verrucomicrobia in the NW group and decreased Firmicutes in the OW group. Meanwhile, the effect of WG on gut microbiota showed gender characteristics, Firmicutes/Bacteroidetes ratio was decreased in women, while Verrucomicrobia abundance was increased in men. The use of WG could improve the microbial composition and promote the growth of beneficial microbes, which may be beneficial to the health of the elderly individuals.
Collapse
Affiliation(s)
- Zeying Cui
- Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Molecular Biology, Hainan Medical University, Haikou, China
| | - Jingtai Li
- Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yuting Zhen
- Key Laboratory of Molecular Biology, Hainan Medical University, Haikou, China
| | - Pingming Fan
- Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- Pingming Fan
| | - Guankui Du
- Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Molecular Biology, Hainan Medical University, Haikou, China
- Department of Biochemistry and Molecular Biology, Hainan Medical University, Haikou, China
- Biotechnology and Biochemistry Laboratory, Hainan Medical University, Haikou, China
- *Correspondence: Guankui Du
| |
Collapse
|
8
|
Liatsos C, Papaefthymiou A, Kyriakos N, Galanopoulos M, Doulberis M, Giakoumis M, Petridou E, Mavrogiannis C, Rokkas T, Kountouras J. Helicobacter pylori, gastric microbiota and gastric cancer relationship: Unrolling the tangle. World J Gastrointest Oncol 2022; 14:959-972. [PMID: 35646287 PMCID: PMC9124990 DOI: 10.4251/wjgo.v14.i5.959] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/12/2021] [Accepted: 04/09/2022] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori infection (Hp-I) represents a typical microbial agent intervening in the complex mechanisms of gastric homeostasis by disturbing the balance between the host gastric microbiota and mucosa-related factors, leading to inflammatory changes, dysbiosis and eventually gastric cancer. The normal gastric microbiota shows diversity, with Proteobacteria [Helicobacter pylori (H. pylori) belongs to this family], Firmicutes, Actinobacteria, Bacteroides and Fusobacteria being the most abundant phyla. Most studies indicate that H. pylori has inhibitory effects on the colonization of other bacteria, harboring a lower diversity of them in the stomach. When comparing the healthy with the diseased stomach, there is a change in the composition of the gastric microbiome with increasing abundance of H. pylori (where present) in the gastritis stage, while as the gastric carcinogenesis cascade progresses to gastric cancer, the oral and intestinal-type pathogenic microbial strains predominate. Hp-I creates a premalignant environment of atrophy and intestinal metaplasia and the subsequent alteration in gastric microbiota seems to play a crucial role in gastric tumorigenesis itself. Successful H. pylori eradication is suggested to restore gastric microbiota, at least in primary stages. It is more than clear that Hp-I, gastric microbiota and gastric cancer constitute a challenging tangle and the strong interaction between them makes it difficult to unroll. Future studies are considered of crucial importance to test the complex interaction on the modulation of the gastric microbiota by H. pylori as well as on the relationships between the gastric microbiota and gastric carcinogenesis.
Collapse
Affiliation(s)
- Christos Liatsos
- Department of Gastroenterology, 401 General Military Hospital of Athens, Athens 11525, Greece
| | - Apostolis Papaefthymiou
- Department of Gastroenterology, 401 General Military Hospital of Athens, Athens 11525, Greece
- Gastroenterology, University Hospital of Larissa, Larissa 41336, Greece
| | - Nikolaos Kyriakos
- Department of Gastroenterology, 401 General Military Hospital of Athens, Athens 11525, Greece
| | - Michail Galanopoulos
- Department of Gastroenterology, 401 General Military Hospital of Athens, Athens 11525, Greece
| | - Michael Doulberis
- Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, Aarau 1234, Switzerland
| | - Marios Giakoumis
- Department of Gastroenterology, 401 General Military Hospital of Athens, Athens 11525, Greece
| | - Evangelia Petridou
- Department of Microbiology, “Agia Sofia” Paediatric Hospital, Goudi, Athens 11527, Greece
| | - Christos Mavrogiannis
- Gastrointestinal and Liver Unit, Faculty of Nursing, Kifissia General and Oncology Hospital, Kaliftaki, N.Kifisia 14564, Greece
| | - Theodore Rokkas
- Gastroenterological Clinic, Henry Dunant Hospital, Athens 11525, Greece
| | - Jannis Kountouras
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki 41336, Macedonia, Greece
| |
Collapse
|
9
|
Shang J, Yang S, Meng X. Correlations between oligosaccharides in breast milk and the composition of the gut microbiome in breastfed infants. J Dairy Sci 2022; 105:4818-4828. [DOI: 10.3168/jds.2021-20928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 02/16/2022] [Indexed: 11/19/2022]
|
10
|
Milk Formula Diet Alters Bacterial and Host Protein Profile in Comparison to Human Milk Diet in Neonatal Piglet Model. Nutrients 2021; 13:nu13113718. [PMID: 34835974 PMCID: PMC8618976 DOI: 10.3390/nu13113718] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 12/25/2022] Open
Abstract
The metaproteome profiling of cecal contents collected from neonatal piglets fed pasteurized human milk (HM) or a dairy-based infant formula (MF) from postnatal day (PND) 2 to 21 were assessed. At PND 21, a subset of piglets from each group (n = 11/group) were euthanized, and cecal contents were collected for further metaproteome analysis. Cecal microbiota composition showed predominantly more Firmicutes phyla and Lachnospiraceae family in the lumen of cecum of HM-fed piglets in comparison to the MF-fed group. Ruminococcus gnavus was the most abundant species from the Firmicutes phyla in the cecal contents of the HM-fed piglets at 21 days of age. A greater number of expressed proteins were identified in the cecal contents of the HM-fed piglets relative to the MF-fed piglets. Greater abundances of proteins potentially expressed by Bacteroides spp. such as glycoside enzymes were noted in the cecal lumen of HM-fed piglets relative to the MF. Additionally, lyases associated with Lachnospiraceae family were abundant in the cecum of the HM group relative to the MF group. Overall, our findings indicate that neonatal diet impacts the gut bacterial taxa and microbial proteins prior to weaning. The metaproteomics data were deposited into PRIDE, PXD025432 and 10.6019/PXD025432.
Collapse
|
11
|
Ferreira RF, Blees T, Shakeri F, Buness A, Sylvester M, Savoini G, Agazzi A, Mrljak V, Sauerwein H. Comparative proteome profiling in exosomes derived from porcine colostrum versus mature milk reveals distinct functional proteomes. J Proteomics 2021; 249:104338. [PMID: 34343709 DOI: 10.1016/j.jprot.2021.104338] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/17/2021] [Accepted: 07/19/2021] [Indexed: 11/24/2022]
Abstract
Exosomes are membranous vesicles of endocytic origin, recently been considered as major players in cell-cell communication. Milk is highly complex, and diverse biocomponents provide adequate nutrition, transfer immunity, and promote adequate neonate development. Milk exosomes are suggested to have a key role in these processes, yet to be further explored, and the alteration of the exosomes' cargo in different stages of lactation stages is important for understanding the factors relevant in nursing and also for improving milk replacer products both for humans and animals. We isolated exosomes from porcine milk in different lactation stages and analyzed their content using a TMT-based high-resolution quantitative proteomic approach. Exosomes were isolated using ultracentrifugation coupled with size exclusion chromatography to enrich milk-derived exosomes in samples obtained at day 0, 7, and 14 after parturition, and characterized by nanoparticle tracking analysis, transmission electron microscopy, and Western blotting. Quantitative proteomics analysis revealed different proteome profiles for colostrum exosomes and milk exosomes. The functional analysis highlighted pathways related to the regulation of homeostasis to be upregulated in colostrum exosomes, and pathways such as endothelial cell development and lipid metabolism to be upregulated in mature milk exosomes. This study endorses the importance of exosomes as active biocomponents of milk and provides knowledge for future studies exploring their role in the regulation of immunity and growth of the newborn. SIGNIFICANCE: The identified functional proteome and protein-protein interaction networks identified in our study help to elucidate the role of milk exosomes in different lactation periods. The results generated herein are of relevance for the basic understanding of their impact on the infant's development but also for bringing forward the manufacturing of milk replacers.
Collapse
Affiliation(s)
- Rafaela Furioso Ferreira
- Institute of Animal Science, Physiology Unit, University of Bonn, Bonn, Germany; Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia.
| | - Thomas Blees
- Institute of Animal Science, Physiology Unit, University of Bonn, Bonn, Germany
| | - Farhad Shakeri
- Institute for Medical Biometry, Informatics and Epidemiology, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany; Institute for Genomic Statistics and Bioinformatics, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany; Core Unit for Bioinformatics Analysis, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Andreas Buness
- Institute for Medical Biometry, Informatics and Epidemiology, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany; Institute for Genomic Statistics and Bioinformatics, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany; Core Unit for Bioinformatics Analysis, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Marc Sylvester
- Institute of Biochemistry and Molecular Biology, Core Facility Mass Spectrometry, Medical Faculty, University of Bonn, Bonn, Germany
| | - Giovanni Savoini
- Department of Health, Animal Science and Food Safety 'Carlo Cantoni' (VESPA), Università degli Studi di Milano, Lodi, Italy
| | - Alessandro Agazzi
- Department of Health, Animal Science and Food Safety 'Carlo Cantoni' (VESPA), Università degli Studi di Milano, Lodi, Italy
| | - Vladimir Mrljak
- Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Helga Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, Bonn, Germany
| |
Collapse
|
12
|
Wang Q, Luo Y, Chaudhuri KR, Reynolds R, Tan EK, Pettersson S. The role of gut dysbiosis in Parkinson's disease: mechanistic insights andtherapeutic options. Brain 2021; 144:2571-2593. [PMID: 33856024 DOI: 10.1093/brain/awab156] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/23/2021] [Accepted: 03/23/2021] [Indexed: 12/02/2022] Open
Abstract
Parkinson's disease is a common neurodegenerative disease in which gastrointestinal symptoms may appear prior to motor symptoms. The gut microbiota of patients with Parkinson's disease shows unique changes, which may be used as early biomarkers of disease. Alteration in gut microbiota composition may be related to the cause or effect of motor or non-motor symptoms, but the specific pathogenic mechanisms are unclear. The gut microbiota and its metabolites have been suggested to be involved in the pathogenesis of Parkinson's disease by regulating neuroinflammation, barrier function and neurotransmitter activity. There is bidirectional communication between the enteric nervous system and the central nervous system, and the microbiota-gut-brain axis may provide a pathway for the transmission of α-synuclein. We highlight recent discoveries and alterations of the gut microbiota in Parkinson's disease, and highlight current mechanistic insights on the microbiota-gut-brain axis in disease pathophysiology. We discuss the interactions between production and transmission of α-synuclein and gut inflammation and neuroinflammation. In addition, we also draw attention to diet modification, use of probiotics and prebiotics and fecal microbiota transplantation as potential therapeutic approaches that may lead to a new treatment paradigm for Parkinson's disease.
Collapse
Affiliation(s)
- Qing Wang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Yuqi Luo
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - K Ray Chaudhuri
- Parkinson Foundation International Centre of Excellence at King's College Hospital, and Kings College, Denmark Hill, London, SE5 9RS, UK
| | - Richard Reynolds
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK.,Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore.,Duke-NUS Medical School, Singapore
| | - Sven Pettersson
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore.,Duke-NUS Medical School, Singapore.,LKC School of Medicine, NTU, Singapore.,Sunway University, Department of Medical Sciences, Kuala Lumpur, Malaysia
| |
Collapse
|
13
|
Wang T, Gao L, Yang Z, Wang F, Guo Y, Wang B, Hua R, Shang H, Xu J. Restraint Stress in Hypertensive Rats Activates the Intestinal Macrophages and Reduces Intestinal Barrier Accompanied by Intestinal Flora Dysbiosis. J Inflamm Res 2021; 14:1085-1110. [PMID: 33790622 PMCID: PMC8007621 DOI: 10.2147/jir.s294630] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/22/2021] [Indexed: 12/14/2022] Open
Abstract
Purpose Hypertension (HTN) is a major risk factor for cardiovascular disease. In recent years, there were numerous studies on the function of stress in HTN. However, the gut dysbiosis linked to hypertension in animal models under stress is still incompletely understood. Purpose of this study is to use multiple determination method to determine the juvenile stage intestinal bacteria, cytokines and changes in hormone levels. Methods Four groups of juvenile male spontaneously hypertensive rats (SHRs) and age-matched male Wistar-Kyoto (WKY) rats were randomly selected as control and experimental groups. Rats in the two stress groups were exposed to restraint stress for 3 hours per day for 7 consecutive days. In one day three times in the method of non-invasive type tail-cuff monitoring blood pressure. The detailed mechanism was illuminated based on the intestinal change using immunohistochemical and immunofluorescence staining and the stress-related hormone and inflammation factors were analyzed via ELISA method. The integrity of the epithelial barrier was assessed using FITC/HRP and the expression levels of proteins associated with the tight junction was detected by Western blot. The alteration of stress-related intestinal flora from ileocecal junction and distal colon were also analyzed using its 16S rDNA sequencing. Results The results indicate that acute stress rapidly increases mean arterial pressure which is positive correlation to hormone concentration, especially in SHR-stress group. Meanwhile, stress promoted the enhancement of epithelial permeability accompanied with a reduced expression of the tight junction-related protein and the macrophages (Mφ) aggregation to the lamina propria. There were remarkable significant increase of stress-related hormones and pro-inflammatory factor interleukin (IL)-6 along with a decrease in the diversity of intestinal flora and an imbalance in the F/B ratio. Conclusion Our results reveal that stress accompanied with HTN could significantly disrupt the domino effect between intestinal flora and homeostasis.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of Physiology and Pathophysiology, Basic Medical College, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Lei Gao
- Department of Biomedical Informatics, School of Biomedical Engineering, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Zejun Yang
- Department of Clinical Medicine, Basic Medical College, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Feifei Wang
- Department of Clinical Medicine, Basic Medical College, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Yuexin Guo
- Department of Oral Medicine, Basic Medical College, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Boya Wang
- Eight Program of Clinical Medicine, Peking University Health Science Center, Beijing, 100081, People's Republic of China
| | - Rongxuan Hua
- Department of Clinical Medicine, Basic Medical College, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Hongwei Shang
- Experimental Center for Morphological Research Platform, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Jingdong Xu
- Department of Physiology and Pathophysiology, Basic Medical College, Capital Medical University, Beijing, 100069, People's Republic of China
| |
Collapse
|
14
|
Morozumi M, Izumi H, Shimizu T, Takeda Y. Comparison of isolation methods using commercially available kits for obtaining extracellular vesicles from cow milk. J Dairy Sci 2021; 104:6463-6471. [PMID: 33714584 DOI: 10.3168/jds.2020-19849] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/22/2021] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles (EV) are important for delivering biologically active substances to facilitate cell-to-cell communication. Milk-derived EV are widely known because of their potential for immune enhancement. However, procedures for isolating milk-derived EV have not been fully established. To obtain pure milk-derived EV and accurately reveal their function, such procedures must be established. The aim of the present study was to compare methods using commercially available kits for isolating milk-derived EV. Initially, we investigated procedures to remove casein, which is the major obstacle in determining milk-derived EV purity. We separated whey using centrifugation only, acetic acid precipitation, and EDTA precipitation. Then, we isolated milk-derived EV by ultracentrifugation, membrane affinity column, size exclusion chromatography (SEC), polymer-based isolation, or phosphatidylserine-affinity isolation. Using EV count per milligram of protein, which is a good indicator of purity, we determined that acetic acid precipitation was the best method for removing casein. Using nanoparticle tracking analysis, protein quantity analysis, and RNA quantity analysis, we comprehensively compared each isolation method for its purity and yield. We found that SEC-based qEV column (Izon Science) could collect purer milk-derived EV at higher quantities. Thus, a combination of acetic acid precipitation and qEV can effectively isolate high amounts of pure extracellular vesicles from bovine milk.
Collapse
Affiliation(s)
- Mai Morozumi
- Wellness and Nutrition Science Institute, Morinaga Milk Industry Co. Ltd., 1-83, 5-Chome, Higashihara, Zama-City, Kanagawa Prefecture 252-8583, Japan.
| | - Hirohisa Izumi
- Wellness and Nutrition Science Institute, Morinaga Milk Industry Co. Ltd., 1-83, 5-Chome, Higashihara, Zama-City, Kanagawa Prefecture 252-8583, Japan
| | - Takashi Shimizu
- Wellness and Nutrition Science Institute, Morinaga Milk Industry Co. Ltd., 1-83, 5-Chome, Higashihara, Zama-City, Kanagawa Prefecture 252-8583, Japan
| | - Yasuhiro Takeda
- Wellness and Nutrition Science Institute, Morinaga Milk Industry Co. Ltd., 1-83, 5-Chome, Higashihara, Zama-City, Kanagawa Prefecture 252-8583, Japan
| |
Collapse
|
15
|
Human Milk-Fed Piglets Have a Distinct Small Intestine and Circulatory Metabolome Profile Relative to That of Milk Formula-Fed Piglets. mSystems 2021; 6:6/1/e01376-20. [PMID: 33563783 PMCID: PMC7883546 DOI: 10.1128/msystems.01376-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Exclusive HM feeding for newborns is recommended at least for the first 6 months of life. However, when breastfeeding is not possible, MF is recommended as a substitute. The impact of human milk (HM) feeding compared with cow’s milk formula (MF) feeding on small intestinal and circulatory metabolome patterns has not been fully investigated. Therefore, 2-day-old male piglets were fed HM or MF (n = 26/group) from postnatal day 2 (PND 2) through 21 and were weaned to a solid diet until PND 51. The small intestine (gastrointestinal [GI]) contents, serum, and urine were collected from subsets of piglets at PND 21 and PND 51. Samples were subjected to primary metabolomics analyses at the West Coast Metabolomics Center, UC Davis. The metabolome data assessment and the statistical analyses were performed with MetaboAnalyst software. Compared with MF feeding, at PND 21, HM feeding resulted in a higher abundance of fucose in the jejunum and urine and a greater concentration of myo-inositol in serum. In HM-fed piglets, 1,5-anhydroglucitol was higher in the duodenum, serum, and urine at PND 21. Additionally, the HM group had higher levels of urinary kynurenic acid at PND 21. Correlations between bacterial genera and altered metabolites in ileum revealed that Turicibacter sp. and Campylobacter sp. were positively correlated with maltotriose and panose at PND 21, while ileal Campylobacter sp. was negatively correlated with fumaric acid. At PND 51, no significant metabolites were identified between HM and MF diet groups. The metabolites associated with the neonatal diets may serve as the substrates and signals that contribute to the physiological effects in HM and MF during infancy, with a subset reflecting diet-associated differences in microbial metabolism and ecology. IMPORTANCE Exclusive HM feeding for newborns is recommended at least for the first 6 months of life. However, when breastfeeding is not possible, MF is recommended as a substitute. Due to the challenges associated with sample collection from infants fed HM or MF, their gut metabolism is poorly understood. Thus, an established piglet model from our team was used to determine the metabolite profile in relation to host, diet, and microbiota. The current study is the first to provide novel insights across the small intestine metabolism and its association with circulatory metabolites in the HM group relative to the MF group at the weaning and postweaning period. Data also demonstrate that during the neonatal period, diet, host, and microbial metabolism contribute to the lumen and circulatory metabolite profile. Furthermore, small intestinal lumen metabolome can be tracked in the urine as a biomarker of dietary differences, which would be a useful tool for clinical interventions.
Collapse
|
16
|
Shabbir U, Rubab M, Daliri EBM, Chelliah R, Javed A, Oh DH. Curcumin, Quercetin, Catechins and Metabolic Diseases: The Role of Gut Microbiota. Nutrients 2021; 13:206. [PMID: 33445760 PMCID: PMC7828240 DOI: 10.3390/nu13010206] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 02/07/2023] Open
Abstract
Polyphenols (PPs) are the naturally occurring bioactive components in fruits and vegetables, and they are the most abundant antioxidant in the human diet. Studies are suggesting that ingestion of PPs might be helpful to ameliorate metabolic syndromes that may contribute in the prevention of several chronic disorders like diabetes, obesity, hypertension, and colon cancer. PPs have structural diversity which impacts their bioavailability as they accumulate in the large intestine and are extensively metabolized through gut microbiota (GM). Intestinal microbiota transforms PPs into their metabolites to make them bioactive. Interestingly, not only GM act on PPs to metabolize them but PPs also modulate the composition of GM. Thus, change in GM from pathogenic to beneficial ones may be helpful to ameliorate gut health and associated diseases. However, to overcome the low bioavailability of PPs, various approaches have been developed to improve their solubility and transportation through the gut. In this review, we present evidence supporting the structural changes that occur after metabolic reactions in PPs (curcumin, quercetin, and catechins) and their effect on GM composition that leads to improving overall gut health and helping to ameliorate metabolic disorders.
Collapse
Affiliation(s)
- Umair Shabbir
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon 24341, Korea; (U.S.); (M.R.); (E.B.-M.D.); (R.C.)
| | - Momna Rubab
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon 24341, Korea; (U.S.); (M.R.); (E.B.-M.D.); (R.C.)
| | - Eric Banan-Mwine Daliri
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon 24341, Korea; (U.S.); (M.R.); (E.B.-M.D.); (R.C.)
| | - Ramachandran Chelliah
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon 24341, Korea; (U.S.); (M.R.); (E.B.-M.D.); (R.C.)
| | - Ahsan Javed
- Department of Food Science & Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Korea;
| | - Deog-Hwan Oh
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon 24341, Korea; (U.S.); (M.R.); (E.B.-M.D.); (R.C.)
| |
Collapse
|
17
|
Zhou X, Zhang Z, Huang F, Yang C, Huang Q. In Vitro Digestion and Fermentation by Human Fecal Microbiota of Polysaccharides from Flaxseed. Molecules 2020; 25:E4354. [PMID: 32977374 PMCID: PMC7582239 DOI: 10.3390/molecules25194354] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/07/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023] Open
Abstract
The digestion of flaxseed polysaccharides (FSP) in simulated saliva, gastric and small intestine conditions was assessed, as well as in vitro fermentation of FSP by human gut microbiota. FSP was not degraded in the simulated digestive systems (there was no change in molecular weight or content of reducing sugars), indicating that ingested FSP would reach the large intestine intact. Changes in carbohydrate content, reducing sugars and culture pH suggested that FSP could be broken down and used by gut microbiota. FSP modulated the composition and structure of the gut microbiota by altering the Firmicutes/Bacteroidetes ratio and increasing the relative abundances of Prevotella, Phascolarctobacterium, Clostridium and Megamonas, which can degrade polysaccharides. Meanwhile, FSP fermentation increased the concentration of short-chain fatty acids, especially propionic and butyric acids. Our results indicate that FSP might be developed as a functional food that benefits gut health.
Collapse
Affiliation(s)
| | | | | | - Chen Yang
- Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Key Laboratory of Oilseeds Processing, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Ministry of Agriculture and Rural affairs, No. 2 Xudong 2nd Road, Wuhan 430062, China; (X.Z.); (Z.Z.); (F.H.); (Q.H.)
| | | |
Collapse
|
18
|
Guo M, Liu G, Chen J, Ma J, Lin J, Fu Y, Fan G, Lee SMY, Zhang L. Dynamics of bacteriophages in gut of giant pandas reveal a potential regulation of dietary intake on bacteriophage composition. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 734:139424. [PMID: 32464399 DOI: 10.1016/j.scitotenv.2020.139424] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/09/2020] [Accepted: 05/12/2020] [Indexed: 06/11/2023]
Abstract
Fecal samples of cubs and adults of giant pandas were examined to determine the effects of diets on the diversity and dynamics of gut bacteriophages. Enterobacteria phage, Salmonella phage, Escherichia phage, Shigella phage, Klebsiella phage, and Lactococcus phage were found to be dominant in both cub and adult samples. Citrobacter phage, Cronobacter phage, Pectobacterium phage, Erwinia phage, Dickeya phage, Erwinia phage, Enterococcus phage, and Pseudomonas phage were more abundant in adults, while Lactococcus phage, Streptococcus phage, Lactobacillus phage, and Leuconostoc phage were more abundant in cubs. The abundance and diversity of the majority of phage species were increased in pandas with age. There was an increase in the abundance of Pectobacterium phage and a decrease in the abundance of Lactobacillus phage, Leuconostoc phage, Bacillus phage, and Streptococcus phage in adults. As cubs and adults of giant pandas have different dietary habits, these observations suggest a significant effect of diets on the composition and abundance of gut bacteriophages in giant pandas.
Collapse
Affiliation(s)
- Min Guo
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Guangdong Institute of Applied Biological Resources, Guangdong Academy of Science, Guangzhou, China; State Key Laboratory of Quality Research of Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Guilin Liu
- BGI-Qingdao, BGI-Shenzhen, Qingdao, China
| | - Jianwei Chen
- BGI-Qingdao, BGI-Shenzhen, Qingdao, China; Synthetic Biology Technology Innovation Center of Shandong Province, Qingdao, China
| | | | | | - Ying Fu
- Faculty of Science and Technology, University of Macau, Macao, China
| | - Guangyi Fan
- State Key Laboratory of Quality Research of Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China; BGI-Qingdao, BGI-Shenzhen, Qingdao, China; BGI-Shenzhen, Shenzhen, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research of Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Libiao Zhang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Guangdong Institute of Applied Biological Resources, Guangdong Academy of Science, Guangzhou, China.
| |
Collapse
|
19
|
Yu Y, Lu J, Oliphant K, Gupta N, Claud K, Lu L. Maternal administration of probiotics promotes gut development in mouse offsprings. PLoS One 2020; 15:e0237182. [PMID: 32764797 PMCID: PMC7413491 DOI: 10.1371/journal.pone.0237182] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/21/2020] [Indexed: 12/22/2022] Open
Abstract
Necrotizing enterocolitis is the most common gastrointestinal disorder in premature neonates. This disease is characterized by massive epithelial necrosis, gut barrier dysfunction and improper mucosal defense development. Studies have shown that probiotic administration can decrease NEC incidence and mortality. The proposed mechanisms of probiotics for the prevention of NEC are: promotion of intestinal development; improved barrier function through decreased apoptosis and improved mucin production; decreased expression of proinflammatory cytokines IL6, IL8, and TNFα, and modulation of microbiota dysbiosis in preterm infants. However, reported sepsis in the immunocompromised preterm host has deterred routine prophylactic administration of probiotics in the neonatal intensive care unit. We hypothesize that maternal administration of probiotics to pregnant mouse dams can recapitulate the beneficial effects observed in neonates fed with probiotics directly. We exposed pregnant mice to the probiotics and monitored the changes in the developing intestines of the offspring. Pregnant mice were fed daily with the probiotics Lactobacillus acidophilus and Bifidobacterium infantis (LB) from embryonic day15 to 2-week-old postnatally. Intraperitoneal administration of IL-1β in the pups was used to model proinflammatory insults. Sera were collected at 2 weeks of age and evaluated for inflammatory cytokines by enzyme-linked-immunosorbent-assay and gut permeability by Fluorescein isothiocyanate-dextran tracer assay. Ileal tissues were collected for the evaluation of apoptosis and proliferation of the intestinal epithelium; as well as mucin and tight junction integrity at mucosal surface by immunofluorescent staining. We find that maternal LB exposure facilitated intestinal epithelial cell differentiation, prevented loss of mucin and preserved the intestinal integrity and barrier function and decreased serum levels of IL-1β, TNF-α and IL-6 in the preweaned offsprings. in LB exposed pups. We demonstrate that maternal probiotic supplementation promotes gut maturation in developing offspring. This is potentially a safe alternative therapy to induce intestinal maturation and prevent prematurity-associated neonatal disorders.
Collapse
Affiliation(s)
- Yueyue Yu
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Jing Lu
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Kaitlyn Oliphant
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Nikhilesh Gupta
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Katerina Claud
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Lei Lu
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
20
|
Elolimy AA, Washam C, Byrum S, Chen C, Dawson H, Bowlin AK, Randolph CE, Saraf MK, Yeruva L. Formula Diet Alters the Ileal Metagenome and Transcriptome at Weaning and during the Postweaning Period in a Porcine Model. mSystems 2020; 5:e00457-20. [PMID: 32753508 PMCID: PMC7406227 DOI: 10.1128/msystems.00457-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/21/2020] [Indexed: 01/05/2023] Open
Abstract
Exclusive breastfeeding impacts the intestinal microbiome and is associated with a better immune function than is seen with milk formula (MF) feeding in infants and yet with mechanisms poorly defined. The porcine model was used to evaluate the impact of MF on ileum microbial communities and gene expression relative to human milk (HM)-fed piglets. Fifty-two Dutch Landrace male piglets were fed an isocaloric diet of either HM (n = 26) or MF (n = 26) from day 2 through day 21 of age and weaned to a solid diet until day 51. Eleven piglets from each group were euthanized at day 21, while the remaining piglets (HM, n = 15; MF, n = 15) were euthanized at day 51 to collect ileal epithelium (EP) scrapings and ileal (IL) tissues. The epithelial mucosa was subjected to shotgun metagenome sequencing, and EP and IL tissues were used for transcriptome analysis. On day 21, transcriptome data revealed that the levels of pathways involved in inflammation and apoptosis were significantly higher in MF piglets than in HM piglets, whereas the levels of tight junctions and pathogen detection systems were lower in MF piglets than in HM piglets. The MF impacts on the small intestine were maintained over the postweaning period (day 51) as indicated by higher levels of Dialister invisus bacteria and higher levels of expression of genes associated with inflammation and apoptosis pathways relative to HM group. The current study demonstrated that MF might impact local intestinal inflammation, apoptosis, and tight junctions and might suppress pathogen recognition in the small intestine compared with HM.IMPORTANCE Exclusive human milk (HM) breastfeeding for the first 6 months of age in infants is recommended to improve health outcomes during early life and beyond. When women are unable to provide sufficient HM, milk formula (MF) is often recommended as a complementary or alternative source of nutrition. Previous studies in piglets demonstrated that MF alters the gut microbiome and induces inflammatory cytokine production. The links between MF feeding, gut microbiome, and inflammation status are unclear due to challenges associated with the collection of intestinal samples from human infants. The current report provides the first insight into MF-microbiome-inflammation connections in the small intestine compared with HM feeding using a porcine model. The present results showed that, compared with HM, MF might impact immune function through the induction of ileal inflammation, apoptosis, and tight junction disruptions and likely compromised immune defense against pathogen detection in the small intestine relative to piglets that were fed HM.
Collapse
Affiliation(s)
- Ahmed A Elolimy
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Charity Washam
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Stephanie Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Celine Chen
- Diet, Genomics & Immunology Laboratory, USDA-ARS Beltsville Human Nutrition Research Center, Beltsville, Maryland, USA
| | - Harry Dawson
- Diet, Genomics & Immunology Laboratory, USDA-ARS Beltsville Human Nutrition Research Center, Beltsville, Maryland, USA
| | - Anne K Bowlin
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | | | - Manish K Saraf
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Laxmi Yeruva
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Arkansas Children's Research Institute, Little Rock, Arkansas, USA
| |
Collapse
|
21
|
Ku HJ, Kim YT, Lee JH. Microbiome Study of Initial Gut Microbiota from Newborn Infants to Children Reveals that Diet Determines Its Compositional Development. J Microbiol Biotechnol 2020; 30:1067-1071. [PMID: 32270658 PMCID: PMC9728344 DOI: 10.4014/jmb.2002.02042] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/03/2020] [Indexed: 12/15/2022]
Abstract
To understand the formation of initial gut microbiota, three initial fecal samples were collected from two groups of two breast milk-fed (BM1) and seven formula milk-fed (FM1) infants, and the compositional changes in gut microbiota were determined using metagenomics. Compositional change analysis during week one showed that Bifidobacterium increased from the first to the third fecal samples in the BM1 group (1.3% to 35.1%), while Klebsiella and Serratia were detected in the third fecal sample of the FM1 group (4.4% and 34.2%, respectively), suggesting the beneficial effect of breast milk intake. To further understand the compositional changes during progression from infancy to childhood (i.e., from three weeks to five years of age), additional fecal samples were collected from four groups of two breast milk-fed infants (BM2), one formula milk-fed toddler (FM2), three weaning food-fed toddlers (WF), and three solid food-fed children (SF). Subsequent compositional change analysis and principal coordinates analysis (PCoA) revealed that the composition of the gut microbiota changed from an infant-like composition to an adult-like one in conjunction with dietary changes. Interestingly, overall gut microbiota composition analyses during the period of progression from infancy to childhood suggested increasing complexity of gut microbiota as well as emergence of a new species of bacteria capable of digesting complex carbohydrates in WF and SF groups, substantiating that diet type is a key factor in determining the composition of gut microbiota. Consequently, this study may be useful as a guide to understanding the development of initial gut microbiota based on diet.
Collapse
Affiliation(s)
- Hye-Jin Ku
- Department of Food Science and Biotechnology, Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea
| | - You-Tae Kim
- Department of Food Science and Biotechnology, Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Ju-Hoon Lee
- Department of Food Science and Biotechnology, Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea,Corresponding author Phone: +82-31-201-3483 Fax: +82-31-204-8116 E-mail:
| |
Collapse
|
22
|
Cioffi CC, Tavalire HF, Neiderhiser JM, Bohannan B, Leve LD. History of breastfeeding but not mode of delivery shapes the gut microbiome in childhood. PLoS One 2020; 15:e0235223. [PMID: 32614839 PMCID: PMC7332026 DOI: 10.1371/journal.pone.0235223] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 06/10/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The naïve neonatal gut is sensitive to early life experiences. Events during this critical developmental window may have life-long impacts on the gut microbiota. Two experiences that have been associated with variation in the gut microbiome in infancy are mode of delivery and feeding practices (eg, breastfeeding). It remains unclear whether these early experiences are responsible for microbial differences beyond toddlerhood. AIMS Our study examined whether mode of delivery and infant feeding practices are associated with differences in the child and adolescent microbiome. DESIGN, SUBJECTS, MEASURES We used an adoption-sibling design to compare genetically related siblings who were reared together or apart. Gut microbiome samples were collected from 73 children (M = 11 years, SD = 3 years, range = 3-18 years). Parents reported on child breastfeeding history, age, sex, height, and weight. Mode of delivery was collected through medical records and phone interviews. RESULTS Negative binomial mixed effects models were used to identify whether mode of delivery and feeding practices were related to differences in phylum and genus-level abundance of bacteria found in the gut of child participants. Covariates included age, sex, and body mass index. Genetic relatedness and rearing environment were accounted for as random effects. We observed a significant association between lack of breastfeeding during infancy and a greater number of the genus Bacteroides in stool in childhood and adolescence. CONCLUSION The absence of breastfeeding may impart lasting effects on the gut microbiome well into childhood.
Collapse
Affiliation(s)
| | | | - Jenae M. Neiderhiser
- The Pennsylvania State University, University Park, PA, United States of America
| | | | - Leslie D. Leve
- University of Oregon, Eugene, OR, United States of America
| |
Collapse
|
23
|
Zhang Y, Jin S, Wang J, Zhang L, Mu Y, Huang K, Zhao B, Zhang K, Cui Y, Li S. Variations in early gut microbiome are associated with childhood eczema. FEMS Microbiol Lett 2020; 366:5376496. [PMID: 30860574 DOI: 10.1093/femsle/fnz020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 03/11/2019] [Indexed: 12/28/2022] Open
Abstract
We assessed the relationship between gut microbiome profile and childhood eczema in 172 subjects (age < 3 years, healthy group N = 123, eczema group N = 49) utilizing 16S rRNA gene sequencing. Lower relative abundance of Bifidobacterium was shown to be associated with childhood eczema. Considering that developmental and environmental factors could modify the state of children's gut microbiome, we divided the samples into four age groups: 0-0.5 years, 0.5-1 years, 1-2 years and 2-3 years for farther analyses. Data revealed significant inter-group differences between healthy and eczema samples in all age groups, and decreased microbial diversity was most significantly found in children with eczema of age 2-3 years old. Decreased abundance of Bifidobacterium was a major finding in eczema groups from 0.5-3 years compared to the age matched healthy controls, but not significant in children younger than 6 month old. Of note, Bifidobacterium operational taxonomic units were identified by Random Forest with highly predictive power of 0.83 (AUC = 0.83) in ROC analysis, which also confirmed its role as a key genus that is associated with eczema. To verify the sequencing results, we performed quantitative polymerase chain reaction of Bifidobacterium and Bacteroides in the same cohort, and in a new eczema cohort (N = 57) for validation. Significantly, lower Bifidobacterium quantities were found in both eczema groups with an age range of 0.5-3 years. These results suggest variations in early gut microbiome are associated with childhood eczema.
Collapse
Affiliation(s)
- Yu Zhang
- Coyote Diagnostics Lab (Beijing) Co., Ltd., Beijing, China
| | - Shujuan Jin
- Coyote Diagnostics Lab (Beijing) Co., Ltd., Beijing, China
| | - Jingjing Wang
- Coyote Diagnostics Lab (Beijing) Co., Ltd., Beijing, China
| | - Lanying Zhang
- Coyote Diagnostics Lab (Beijing) Co., Ltd., Beijing, China
| | - Yu Mu
- Prediatric Department, No.16. Taiyanggong Middel Rd Guanjie Building Chaoyang District, Beijing, China, 100028
| | - Kefei Huang
- Prediatric Department, No.16. Taiyanggong Middel Rd Guanjie Building Chaoyang District, Beijing, China, 100028
| | - Bo Zhao
- Coyote Diagnostics Lab (Beijing) Co., Ltd., Beijing, China
| | - Kejian Zhang
- Coyote Diagnostics Lab (Beijing) Co., Ltd., Beijing, China
| | - Yutao Cui
- Prediatric Department, No.16. Taiyanggong Middel Rd Guanjie Building Chaoyang District, Beijing, China, 100028
| | - Sabrina Li
- Coyote Diagnostics Lab (Beijing) Co., Ltd., Beijing, China.,Coyote Bioscience (Beijing) Co., Ltd., Beijing, China
| |
Collapse
|
24
|
Capuco A, Urits I, Hasoon J, Chun R, Gerald B, Wang JK, Ngo AL, Simopoulos T, Kaye AD, Colontonio MM, Parker-Actlis TQ, Fuller MC, Viswanath O. Gut Microbiome Dysbiosis and Depression: a Comprehensive Review. Curr Pain Headache Rep 2020; 24:36. [DOI: 10.1007/s11916-020-00871-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
25
|
Brink LR, Mercer KE, Piccolo BD, Chintapalli SV, Elolimy A, Bowlin AK, Matazel KS, Pack L, Adams SH, Shankar K, Badger TM, Andres A, Yeruva L. Neonatal diet alters fecal microbiota and metabolome profiles at different ages in infants fed breast milk or formula. Am J Clin Nutr 2020; 111:1190-1202. [PMID: 32330237 PMCID: PMC7266684 DOI: 10.1093/ajcn/nqaa076] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 03/26/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Neonatal diet has a large influence on child health and might modulate changes in fecal microbiota and metabolites. OBJECTIVES The aim is to investigate fecal microbiota and metabolites at different ages in infants who were breastfed (BF), received dairy-based milk formula (MF), or received soy-based formula (SF). METHODS Fecal samples were collected at 3 (n = 16, 12, and 14, respectively), 6 (n = 20, 19, and 15, respectively), 9 (n = 12, 11, and 12, respectively), and 12 mo (n = 14, 14, and 15, respectively) for BF, MF, and SF infants. Infants that breastfed until 9 mo and switched to formula were considered as no longer breastfeeding at 12 mo. Microbiota data were obtained using 16S ribosomal RNA sequencing. Untargeted metabolomics was conducted using a Q-Exactive Hybrid Quadrupole-Orbitrap mass spectrometer. The data were analyzed using R (version 3.6.0) within the RStudio (version 1.1.463) platform. RESULTS At 3, 6, and 9 mo of age BF infants had the lowest α-diversity, SF infants had the highest diversity, and MF was intermediate. Bifidobacterium was 2.6- to 5-fold lower in SF relative to BF infants through 1 y of life. An unidentified genus from Ruminococcaceae higher in the SF (2%) than in the MF (0.4%) and BF (0.08%) infants at 3 mo of age was observed. In BF infants higher levels of butyric acid, d-sphingosine, kynurenic acid, indole-3-lactic acid, indole-3-acetic acid, and betaine were observed than in MF and SF infants. At 3 mo Ruminococcaceae was positively correlated to azelaic, gentisic, isocitric, sebacic, and syringic acids. At 6 mo Oscillospira was negatively correlated with 3-hydroxybutyric-acid, hydroxy-hydrocinnamic acid, and betaine whereas Bifidobacterium was negatively associated with 5-hydroxytryptamine. At 12 mo of age, Lachnospiraceae was negatively associated with hydroxyphenyllactic acid. CONCLUSIONS Infant diet has a large impact on the fecal microbiome and metabolome in the first year of life.This study was registered at clinicaltrials.gov as NCT00616395.
Collapse
Affiliation(s)
- Lauren R Brink
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kelly E Mercer
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Brian D Piccolo
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ahmed Elolimy
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Anne K Bowlin
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Katelin S Matazel
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Lindsay Pack
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sean H Adams
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kartik Shankar
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Thomas M Badger
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Aline Andres
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Laxmi Yeruva
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Arkansas Children's Research Institute, Little Rock, AR, USA
| |
Collapse
|
26
|
Osakunor DNM, Munk P, Mduluza T, Petersen TN, Brinch C, Ivens A, Chimponda T, Amanfo SA, Murray J, Woolhouse MEJ, Aarestrup FM, Mutapi F. The gut microbiome but not the resistome is associated with urogenital schistosomiasis in preschool-aged children. Commun Biol 2020; 3:155. [PMID: 32242065 PMCID: PMC7118151 DOI: 10.1038/s42003-020-0859-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/25/2020] [Indexed: 02/07/2023] Open
Abstract
Helminth parasites have been shown to have systemic effects in the host. Using shotgun metagenomic sequencing, we characterise the gut microbiome and resistome of 113 Zimbabwean preschool-aged children (1-5 years). We test the hypothesis that infection with the human helminth parasite, Schistosoma haematobium, is associated with changes in gut microbial and antimicrobial resistance gene abundance/diversity. Here, we show that bacteria phyla Bacteroidetes, Firmicutes, Proteobacteria, and fungi phyla Ascomycota, Microsporidia, Zoopagomycota dominate the microbiome. The abundance of Proteobacteria, Ascomycota, and Basidiomycota differ between schistosome-infected versus uninfected children. Specifically, infection is associated with increases in Pseudomonas, Stenotrophomonas, Derxia, Thalassospira, Aspergillus, Tricholoma, and Periglandula, with a decrease in Azospirillum. We find 262 AMR genes, from 12 functional drug classes, but no association with individual-specific data. To our knowledge, we describe a novel metagenomic dataset of Zimbabwean preschool-aged children, indicating an association between urogenital schistosome infection and changes in the gut microbiome.
Collapse
Affiliation(s)
- Derick N M Osakunor
- Institute of Immunology & Infection Research, University of Edinburgh, Ashworth Laboratories, King's Buildings, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK.
| | - Patrick Munk
- Research Group for Genomic Epidemiology, National Food Institute, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark
| | - Takafira Mduluza
- Biochemistry Department, University of Zimbabwe, P.O. Box MP167, Mount Pleasant, Harare, Zimbabwe
| | - Thomas N Petersen
- Research Group for Genomic Epidemiology, National Food Institute, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark
| | - Christian Brinch
- Research Group for Genomic Epidemiology, National Food Institute, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark
| | - Alasdair Ivens
- Institute of Immunology & Infection Research, University of Edinburgh, Ashworth Laboratories, King's Buildings, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
| | - Theresa Chimponda
- Biochemistry Department, University of Zimbabwe, P.O. Box MP167, Mount Pleasant, Harare, Zimbabwe
| | - Seth A Amanfo
- Usher Institute of Population Health Sciences & Informatics, University of Edinburgh, Ashworth Laboratories, Kings Buildings, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
- NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), University of Edinburgh, Ashworth Laboratories, King's Buildings, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
| | - Janice Murray
- Institute of Immunology & Infection Research, University of Edinburgh, Ashworth Laboratories, King's Buildings, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
- NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), University of Edinburgh, Ashworth Laboratories, King's Buildings, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
| | - Mark E J Woolhouse
- Usher Institute of Population Health Sciences & Informatics, University of Edinburgh, Ashworth Laboratories, Kings Buildings, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
- NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), University of Edinburgh, Ashworth Laboratories, King's Buildings, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
| | - Frank M Aarestrup
- Research Group for Genomic Epidemiology, National Food Institute, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark
| | - Francisca Mutapi
- Institute of Immunology & Infection Research, University of Edinburgh, Ashworth Laboratories, King's Buildings, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
- NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), University of Edinburgh, Ashworth Laboratories, King's Buildings, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
| |
Collapse
|
27
|
Abstract
The human gut microbiome partakes in a bidirectional communication pathway with the central nervous system (CNS), named the microbiota–gut–brain axis. The microbiota–gut–brain axis is believed to modulate various central processes through the vagus nerve as well as production of microbial metabolites and immune mediators which trigger changes in neurotransmission, neuroinflammation, and behavior. Little is understood about the utilization of microbiome manipulation to treat disease. Though studies exploring the role of the microbiome in various disease processes have shown promise, mechanisms remain unclear and evidence-based treatments for most illnesses have not yet been developed. The animal studies reviewed here offer an excellent array of basic science research that continues to clarify mechanisms by which the microbiome may affect mental health. More evidence is needed, particularly as it relates to translating this work to human subjects. The studies presented in this paper largely demonstrate encouraging results in the treatment of depression. Limitations include small sample sizes and heterogeneous methodology. The exact mechanism by which the gut microbiota causes or alters neuropsychiatric disease states is not fully understood. In this review, we focus on recent studies investigating the relationship between gut microbiome dysbiosis and the pathogenesis of depression. This article is based on previously conducted studies and does not contain any studies with human participants or animals performed by any of the authors.
Collapse
|
28
|
Brink LR, Matazel K, Piccolo BD, Bowlin AK, Chintapalli SV, Shankar K, Yeruva L. Neonatal Diet Impacts Bioregional Microbiota Composition in Piglets Fed Human Breast Milk or Infant Formula. J Nutr 2019; 149:2236-2246. [PMID: 31373372 PMCID: PMC6888031 DOI: 10.1093/jn/nxz170] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/07/2019] [Accepted: 06/26/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Early infant diet influences postnatal gut microbial development, which in turn can modulate the developing immune system. OBJECTIVES The aim of this study was to characterize diet-specific bioregional microbiota differences in piglets fed either human breast milk (HM) or infant formula. METHODS Male piglets (White Dutch Landrace Duroc) were raised on HM or cow milk formula (MF) from postnatal day (PND) 2 to PND 21 and weaned to an ad libitum diet until PND 51. Piglets were euthanized on either PND 21 or PND 51, and the gastrointestinal contents were collected for 16s RNA sequencing. Data were analyzed using the Quantitative Insight into Microbial Ecology. Diversity measurements (Chao1 and Shannon) and the Wald test were used to determine relative abundance. RESULTS At PND 21, the ileal luminal region of HM-fed piglets showed lower Chao1 operational taxonomic unit diversity, while Shannon diversity was lower in cecal, proximal colon (PC), and distal colon (DC) luminal regions, relative to MF-fed piglets. In addition, at PND 51, the HM-fed piglets had lower genera diversity within the jejunum, ileum, PC, and DC luminal regions, relative to MF-fed piglets. At PND 21, Turicibacter was 4- to 5-fold lower in the HM-fed piglets' ileal, cecal, PC, and DC luminal regions, relative to the MF-fed piglets. Campylobacter is 3- to 6-fold higher in HM-fed piglets duodenal, ileal, cecal, PC, and DC luminal regions, in comparison to MF-fed piglets. Furthermore, the large intestine (cecum, PC, and rectum) luminal region of HM-fed piglets showed 4- to 7-fold higher genera that belong to class Bacteroidia, in comparison to MF-fed piglets at PND 21. In addition, at PND 51 distal colon lumen of HM-fed piglets showed 1.5-fold higher genera from class Bacteroidia than the MF-fed piglets. CONCLUSIONS In the large intestinal regions (cecum, PC, and rectum), MF diet alters microbiota composition, relative to HM diet, with sustained effects after weaning from the neonatal diet. These microbiota changes could impact immune system and health outcomes later in life.
Collapse
Affiliation(s)
- Lauren R Brink
- Arkansas Children's Nutrition Center, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Katelin Matazel
- Arkansas Children's Nutrition Center, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Brian D Piccolo
- Arkansas Children's Nutrition Center, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Anne K Bowlin
- Arkansas Children's Nutrition Center, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kartik Shankar
- Arkansas Children's Nutrition Center, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Laxmi Yeruva
- Arkansas Children's Nutrition Center, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Arkansas Children's Research Institute, Little Rock, AR, USA
| |
Collapse
|
29
|
Wang M, Radlowski EC, Li M, Monaco MH, Donovan SM. Feeding Mode, but Not Prebiotics, Affects Colonic Microbiota Composition and Volatile Fatty Acid Concentrations in Sow-Reared, Formula-Fed, and Combination-Fed Piglets. J Nutr 2019; 149:2156-2163. [PMID: 31504685 DOI: 10.1093/jn/nxz183] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/24/2019] [Accepted: 07/15/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Many infants consume both human milk and infant formula (combination-fed); however, little is known about how combination-feeding affects the gut microbiota or prebiotic fermentation compared to formula feeding. OBJECTIVES We investigated the impact of feeding mode and prebiotics on bacterial colonization and volatile fatty acid (VFA) concentrations. METHODS Newborn piglets (Large White and Landrace) were randomly assigned to 5 groups (n = 6/group): formula-fed (FF), formula-fed with prebiotics (FP), sow-reared (SR), combination-fed (CF), and combination-fed with prebiotics (CP). SR piglets remained with the sows 24 h/d. FF and FP were fed formula or formula with galactooligosaccharide and inulin (4 g/L in a 4:1 ratio). CF and CP were sow-reared for 5 d and then rotated between the sow and formula-feeding every 12 h. Ascending colon contents were collected at day 21. The microbiota was analyzed by pyrosequencing and denaturing gradient gel electrophoresis (DGGE). VFAs were determined by gas chromatography. RESULTS Distance-based redundancy analysis of DGGE and pyrosequencing data separated microbiota of FF from CF and SR. CF differed from SR by DGGE, but only a trend (P = 0.09) by pyrosequencing. Bacterial composition of CF was more similar to SR than FF. No bacterial genera in CF significantly differed from SR; however, 9 genera differed between CF and FF, including Lactobacillus, Clostridium XIVa, and Fusobacterium. VFA concentrations were similar between CF and SR, while isovalerate and isobutyrate were 2-fold greater (P < 0.05) in CF than FF. Neither microbiota nor VFA profile was affected by prebiotic supplementation. CONCLUSIONS Microbial colonization patterns and VFA profiles of CF piglets were more similar to SR piglets than FF piglets. Prebiotics did not affect piglet bacterial composition and/or VFA concentrations relative to the main feeding modes (FF and CF). Thus, partial exposure to breast milk can be beneficial for microbiota development of FF neonates.
Collapse
Affiliation(s)
- Mei Wang
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL, USA
| | - Emily C Radlowski
- Department of Nutrition Sciences, Dominican University, River Forest, IL, USA
| | - Min Li
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL, USA
| | - Marcia H Monaco
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL, USA
| | - Sharon M Donovan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL, USA.,Division of Nutritional Sciences, University of Illinois, Urbana, IL, USA
| |
Collapse
|
30
|
Effects of Maternal Supplementation with Rare Earth Elements during Late Gestation and Lactation on Performances, Health, and Fecal Microbiota of the Sows and Their Offspring. Animals (Basel) 2019; 9:ani9100738. [PMID: 31569383 PMCID: PMC6826669 DOI: 10.3390/ani9100738] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 09/12/2019] [Accepted: 09/13/2019] [Indexed: 12/19/2022] Open
Abstract
Simple Summary The immunological and metabolic status of breeding sows directly affect the overall productivity of porcine operations. Especially, maternal health status during the transition from gestation to lactation are important in maintaining health and growth of the suckling piglets. Rare earth elements (REEs) have been considered as a promising natural feed additive and been reported to exert their activity locally within the gastrointestinal tract, including effects on the bacterial microflora and on nutrient utilization. The present study was conducted to explore the effects of dietary maternal REE supplementation during late gestation and lactation on sows and their offspring. After the experiment, we found that maternal REE addition enhanced antioxidant activity and immunity of sows and their suckling piglets. At the same time, REE supplementation during perinatal period improved the reproductivity of the sows as well as the growth of their offspring. Besides, maternal REEs supply altered the intestinal microbiota community and composition of sows as well as their offspring, and Spearman correlation analysis shows that fecal bacteria are associated with the antioxidase, inflammatory factors of the sows and offspring as well as average daily gain of the suckling piglets. In addition, our results suggested that REE supplementation during both gestation and lactation are more beneficial to sows and their offspring than supplementation during only late gestation. This paper holds promise in providing efficient feeding strategies in swine production. Abstract The study was conducted to investigate the effects of maternal supplementation with rare earth elements (REEs) on sows and their offspring. During late gestation, 120 multiparous sows were divided randomly into the control group (Basal diet) and REE-G group (Basal diet supplemented with 200 mg REE/kg). After delivery, REE-G group was further divided into two groups: REE-L- (Change to basal diet during lactation) and REE-L+ group (REE diet all the time). Our results showed that maternal REE supplementation improved the antioxidant and immunity of sows and piglets. Additionally, REE supply during late gestation significantly decreased the coefficient of within-litter variation (CV) in birth weight and increased the weaning weights and the average daily gain (ADG) of piglets. During lactation, the insulin-like growth factor-1 (IGF-1) levels in piglets of REE-L+ group were higher, while no difference between REE-L- and the control group. More beneficial bacteria (Christensenellaceae and Ruminococcaceae) were found in the REE-L+ group while some opportunistic pathogens (Proteobacteria and Campylobacter) were relatively suppressed. Fecal microbiota showed correlation with antioxidase, inflammatory factors, and average daily gain (ADG). Collectively, our findings indicated that REEs added in both gestation and lactation was more conducive to establish a healthier status for sows and their offspring.
Collapse
|
31
|
Influence of Socio-Economic and Psychosocial Profiles on the Human Breast Milk Bacteriome of South African Women. Nutrients 2019; 11:nu11061390. [PMID: 31226873 PMCID: PMC6627120 DOI: 10.3390/nu11061390] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 12/02/2022] Open
Abstract
The human breast milk (HBM) bacteriome is an important, continuous source of microbes to the neonate in early life, playing an important role in shaping the infant’s intestinal bacteriome. Study of the composition of the HBM bacteriome is an emerging area of research, with little information available, particularly from low- and middle-income countries. The aim of this study was to characterize the diversity of bacterial communities in HBM samples collected between 6–10 weeks postpartum from lactating South African women and to study potential influencing factors of the bacteriome. Using 16S rRNA gene sequencing of samples from 554 women, we demonstrated that the HBM bacteriome was largely dominated by the phyla Firmicutes (mean relative abundance: 71.1%) and Actinobacteria (mean relative abundance: 16.4%). The most abundant genera identified from the HBM bacteriome were Streptococcus (mean relative abundance: 48.6%), Staphylococcus (mean relative abundance: 17.8%), Rothia (mean relative abundance: 5.8%), and Corynebacterium (mean relative abundance: 4.3%). “Core” bacterial genera including Corynebacterium, Streptococcus, Staphylococcus, Rothia, Veillonella, Gemella, Acinetobacter, Micrococcus and a genus belonging to the Enterobacteriaceae family were present in 80% of samples. HBM samples were classified, according to their bacteriome, into three major clusters, dominated by the genera Staphylococcus (cluster 1), a combination of Staphylococcus and Streptococcus (cluster 2), and Streptococcus (cluster 3). The cluster groups differed significantly for Shannon and chao1 richness indices. Bacterial interactions were studied using co-occurrence networks with positive associations observed between the abundances of Staphylococcus and Corynebacteria (members of the skin microflora) and between Streptococcus, Rothia, Veillonella, and Gemella (members of the oral microflora). HBM from older mothers had a higher Shannon diversity index. The study site was associated with differences in HBM bacteriome composition (permutational multivariate analysis of variance using distance matrices (PERMANOVA), p < 0.05). No other tested socio-demographic or psychosocial factors were associated with HBM bacterial composition.
Collapse
|
32
|
Andrews K, Gonzalez A. Contextual risk factors impacting the colonization and development of the intestinal microbiota: Implications for children in low- and middle-income countries. Dev Psychobiol 2019; 61:714-728. [PMID: 30663777 DOI: 10.1002/dev.21823] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 12/09/2018] [Accepted: 12/11/2018] [Indexed: 12/11/2022]
Abstract
Early adversities, such as poverty, maternal stress, and malnutrition, can affect the structure and functioning of the developing brain with implications for biological systems such as the intestinal microbiota. To date, most studies examining the impact of these risk factors on the development and functioning of the intestinal microbiota have primarily been conducted in high-income countries. However, arguably, children in low- and middle-income countries may be at increased risk given cumulative biological and psychosocial adversities during their development. Accumulating evidence in low- and middle-income countries has linked dysbiosis of the intestinal microbiota to child health outcomes such as stunting, malnutrition, and diarrheal diseases characterized by reduced microbial diversity and elevated pathogenic bacteria, which has implications for psychosocial outcomes. This review summarizes empirical findings that highlight the association between risk factors prevalent in low- and middle-income countries and the intestinal microbiota of children. Additionally, we briefly survey the current evidence regarding the effect of nutritional interventions on the microbial composition of children in low- and middle-income countries. We conclude that these empirical studies have the capacity to inform future research investigating the influence of preventive interventions on biological systems by targeting the predominant risk factors faced by children in low- and middle-income countries.
Collapse
Affiliation(s)
- Krysta Andrews
- Neuroscience Graduate Program, McMaster University, Hamilton, Ontario, Canada.,Offord Centre for Child Studies, McMaster University, Hamilton, Ontario, Canada
| | - Andrea Gonzalez
- Offord Centre for Child Studies, McMaster University, Hamilton, Ontario, Canada.,Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
33
|
Osadchiy V, Martin CR, Mayer EA. The Gut-Brain Axis and the Microbiome: Mechanisms and Clinical Implications. Clin Gastroenterol Hepatol 2019; 17:322-332. [PMID: 30292888 PMCID: PMC6999848 DOI: 10.1016/j.cgh.2018.10.002] [Citation(s) in RCA: 311] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 09/26/2018] [Accepted: 10/01/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Based largely on results from preclinical studies, the concept of a brain gut microbiome axis has been established, mediating bidirectional communication between the gut, its microbiome, and the nervous system. Limited data obtained in human beings suggest that alterations in these interactions may play a role in several brain gut disorders. METHODS We reviewed the preclinical and clinical literature related to the topic of brain gut microbiome interactions. RESULTS Well-characterized bidirectional communication channels, involving neural, endocrine, and inflammatory mechanisms, exist between the gut and the brain. Communication through these channels may be modulated by variations in the permeability of the intestinal wall and the blood-brain barrier. Brain gut microbiome interactions are programmed during the first 3 years of life, including the prenatal period, but can be modulated by diet, medications, and stress throughout life. Based on correlational studies, alterations in these interactions have been implicated in the regulation of food intake, obesity, and in irritable bowel syndrome, even though causality remains to be established. CONCLUSIONS Targets within the brain gut microbiome axis have the potential to become targets for novel drug development for brain gut disorders.
Collapse
|
34
|
Miklavcic JJ, Badger TM, Bowlin AK, Matazel KS, Cleves MA, LeRoith T, Saraf MK, Chintapalli SV, Piccolo BD, Shankar K, Yeruva L. Human Breast-Milk Feeding Enhances the Humoral and Cell-Mediated Immune Response in Neonatal Piglets. J Nutr 2018; 148:1860-1870. [PMID: 30247686 PMCID: PMC6209812 DOI: 10.1093/jn/nxy170] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 06/22/2018] [Accepted: 07/03/2018] [Indexed: 12/24/2022] Open
Abstract
Background The benefits of breastfeeding infants are well characterized, including those on the immune system. However, determining the mechanism by which human breast milk (HBM) elicits effects on immune response requires investigation in an appropriate animal model. Objective The primary aim of this study was to develop a novel porcine model and to determine the differential effects of feeding HBM and a commercial milk formula (MF) on immune response and gastrointestinal microbial colonization in a controlled environment. Methods Male piglets were fed HBM (n = 26) or MF (n = 26) from day 2 through day 21. Piglets were vaccinated (n = 9/diet group) with cholera toxin and cholera toxin subunit B (CTB) and tetanus toxoid at 21 d or were fed placebo (n = 6/diet group) and then weaned to a standard solid diet at the age of 21 d. Humoral and cell-mediated immune responses were assessed from blood on days 35 and 48. Immune response was further examined from tissues, including mesenteric lymph nodes (MLNs), Peyer's patches (PPs), and spleen. The colonization of gut microbiota was characterized from feces on days 16 and 49. Results Serum antibody titers in piglets fed HBM were 4-fold higher (P < 0.05) to CTB and 3-fold higher (P < 0.05) to tetanus toxoid compared with piglets fed MF on day 48. Compared with MF, the numbers of immunoglobulin A antibody-producing cells to CTB were 13-fold higher (P < 0.05) in MLNs and 11-fold higher (P < 0.05) in PPs in the HBM diet group on day 51. In addition, significantly increased T cell proliferation was observed in the HBM group relative to the MF group. Furthermore, microbial diversity in the HBM group was lower (P < 0.05) than in the MF group. Conclusions This porcine model appears to be valid for studying the effects of early postnatal diet on immune responses and the gastrointestinal microbiome. Our results lay the groundwork for future studies defining the role of infant diet on microbiota and immune function.
Collapse
Affiliation(s)
- John J Miklavcic
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Thomas M Badger
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Anne K Bowlin
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Katelin S Matazel
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Mario A Cleves
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
- Arkansas Children's Research Institute, Little Rock, AR
| | - Tanya LeRoith
- Department of Biomedical Science and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - Manish K Saraf
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Brian D Piccolo
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Kartik Shankar
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Laxmi Yeruva
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
- Arkansas Children's Research Institute, Little Rock, AR
| |
Collapse
|
35
|
Ginsenosides, catechins, quercetin and gut microbiota: Current evidence of challenging interactions. Food Chem Toxicol 2018; 123:42-49. [PMID: 30336256 DOI: 10.1016/j.fct.2018.10.042] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/08/2018] [Accepted: 10/15/2018] [Indexed: 12/16/2022]
Abstract
Recent studies have shown the role of gut microbiota in favoring the absorption of herbal products and the transformation of their active principles into metabolites endowed with biological activity. This review focuses on the evidence supporting the changes occurring, after metabolic reactions by specific bacteria that colonize the human gut, to ginseng-derived ginsenosides, green tea-derived catechins, and quercetin, this latter being a flavonoid aglycon bound to sugars and abundant in some vegetables and roots. Furthermore, the results of several studies demonstrating the potential beneficial effects of the active metabolites generated by these biotransformations on ginsenosides, catechins and quercetin will be reported.
Collapse
|
36
|
Sharma S, Tripathi P. Gut microbiome and type 2 diabetes: where we are and where to go? J Nutr Biochem 2018; 63:101-108. [PMID: 30366260 DOI: 10.1016/j.jnutbio.2018.10.003] [Citation(s) in RCA: 230] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 09/17/2018] [Accepted: 10/03/2018] [Indexed: 12/22/2022]
Abstract
Type 2 diabetes mellitus (T2D) is a highly prevalent metabolic disorder characterized by an imbalance in blood glucose level, altered lipid profile and high blood pressure. Genetic constituents, high-fat and high-energy dietary habits, and a sedentary lifestyle are three major factors that contribute to high risk of T2D. Several studies have reported gut microbiome dysbiosis as a factor in rapid progression of insulin resistance in T2D that accounts for about 90% of all diabetes cases worldwide. The gut microbiome dysbiosis may reshape intestinal barrier functions and host metabolic and signaling pathways, which are directly or indirectly related to the insulin resistance in T2D. Thousands of the metabolites derived from microbes interact with the epithelial, hepatic and cardiac cell receptors that modulate host physiology. Xenobiotics including dietary components, antibiotics and nonsteroidal anti-inflammatory drugs strongly affect the gut microbial composition and can promote dysbiosis. Any change in the gut microbiota can shift the host metabolism towards increased energy harvest during diabetes and obesity. However, the exact mechanisms behind the dynamics of gut microbes and their impact on host metabolism at the molecular level are yet to be deciphered. We reviewed the published literature for better understanding of the dynamics of gut microbiota, factors that potentially induce gut microbiome dysbiosis and their relation to the progression of T2D. Special emphasis was also given to understand the gut microbiome induced breaching of intestinal barriers and/or tight junctions and their relation to insulin resistance.
Collapse
Affiliation(s)
- Sapna Sharma
- Gene Regulation Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Prabhanshu Tripathi
- Centre for Human Microbial Ecology, Translational Health Science, and Technological Institute, NCR Biotech Science Cluster, 3rd Milestone Gurgaon-Faridabad Expressway, Faridabad, Haryana 121001, India.
| |
Collapse
|
37
|
Zhao J, Yao Y, Li D, Xu H, Wu J, Wen A, Xie M, Ni Q, Zhang M, Peng G, Xu H. Characterization of the Gut Microbiota in Six Geographical Populations of Chinese Rhesus Macaques (Macaca mulatta), Implying an Adaptation to High-Altitude Environment. MICROBIAL ECOLOGY 2018; 76:565-577. [PMID: 29372281 DOI: 10.1007/s00248-018-1146-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 01/09/2018] [Indexed: 05/05/2023]
Abstract
Knowledge about the impact of different geographical environments on rhesus macaque gut microbiota is limited. In this study, we compared the characteristics of gut microbiota in six different Chinese rhesus macaque populations, including Hainan, Nanning, Guizhou, Xichang, Jianchuan and Tibet. Through the composition analysis of operational taxonomic units (OTUs), we found that there were significant differences in the abundance of core overlapping OTUs in the six Chinese groups. Specifically, the Tibet population exhibited the highest gut microbial diversity and the most unique OTUs. Statistically significant differences in the composition of gut microbiota among the six groups at phylum and family level were evident. Specifically, Tibet had higher abundances of Firmicutes and lower abundances of Bacteroidetes than the other geographical groups, and the higher abundance of Firmicutes in the Tibetan group was mainly caused by a significant increase in the family Ruminococcaceae and Christensenellaceae. Phylogenetic investigation of communities by reconstruction of unobserved state analysis showed that the enrichment ratio for environmental information processing and organismal systems was the highest in the Tibet population. Additionally, our results suggested that in the adaptation process of rhesus macaques to different geographical environments, the abundance of the core common flora of the intestinal microbes had undergone varying degree of change and produced new and unique flora, both of which helped to reshape the gut microbiota of rhesus macaques. In particular, this change was more obvious for animals in the high-altitude environments.
Collapse
Affiliation(s)
- Junsong Zhao
- College of Life Science, Sichuan Agricultural University, No. 46, Xinkang Road, Yucheng District, Ya'an, 625014, Sichuan, People's Republic of China
| | - Yongfang Yao
- College of Life Science, Sichuan Agricultural University, No. 46, Xinkang Road, Yucheng District, Ya'an, 625014, Sichuan, People's Republic of China
| | - Diyan Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Huaming Xu
- College of Life Science, Sichuan Agricultural University, No. 46, Xinkang Road, Yucheng District, Ya'an, 625014, Sichuan, People's Republic of China
| | - Jiayun Wu
- College of Life Science, Sichuan Agricultural University, No. 46, Xinkang Road, Yucheng District, Ya'an, 625014, Sichuan, People's Republic of China
| | - Anxiang Wen
- College of Life Science, Sichuan Agricultural University, No. 46, Xinkang Road, Yucheng District, Ya'an, 625014, Sichuan, People's Republic of China
| | - Meng Xie
- College of Life Science, Sichuan Agricultural University, No. 46, Xinkang Road, Yucheng District, Ya'an, 625014, Sichuan, People's Republic of China
| | - Qingyong Ni
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Mingwang Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Guangneng Peng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Huailiang Xu
- College of Life Science, Sichuan Agricultural University, No. 46, Xinkang Road, Yucheng District, Ya'an, 625014, Sichuan, People's Republic of China.
| |
Collapse
|
38
|
Ossa JC, Yáñez D, Valenzuela R, Gallardo P, Lucero Y, Farfán MJ. Intestinal Inflammation in Chilean Infants Fed With Bovine Formula vs. Breast Milk and Its Association With Their Gut Microbiota. Front Cell Infect Microbiol 2018; 8:190. [PMID: 29977866 PMCID: PMC6022179 DOI: 10.3389/fcimb.2018.00190] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 05/17/2018] [Indexed: 12/26/2022] Open
Abstract
Introduction: Compared to bovine formula (BF), breast milk (BM) has unique properties. In the newborn intestine, there is a homeostatic balance between the counterparts of the immune system, which allows a physiological inflammation, modulated by the gut microbiota. Many studies have attempted to understand the effect of BF vs. BM, and the changes in the gut microbiota, but few also focus on intestinal inflammation. Methods: We conducted a cohort study of newborn infants during their first 3 months. In stool samples taken at 1 and 3 months (timepoints T1 and T3), we quantified calprotectin, IL-8 and α1-antitrypsin by ELISA and we evaluated the expression of IL8 and IL1β genes by RT-qPCR. To determine the microbiota composition, the 16S rRNA gene was amplified and sequenced using 454 pyrosequencing. Sequences were clustered into operational taxonomic units (OTUs). Results: In total 15 BM and 10 BF infants were enrolled. In the BM group, we found calprotectin and α1-antitrypsin levels were significantly elevated at T3 compared to T1; no differences were found between T1 and T3 in the BF group. A comparison between the BM and BF groups showed that calprotectin levels at T1 were lower in the BM than the BF group; this difference was not observed at T3. For IL-8 levels, we found no differences between groups. A gene expression analysis of the IL8 and IL1β genes showed that infants from the BF group at T1 have a significantly increased expression of these markers compared to the BM group. Gut microbiota analyses revealed that the phylum Bacteroidetes was higher in BM than BF, whereas Firmicutes were higher in BF. A redundancy analysis and ANOVA showed BM has a community structure statistically different to BF at T1 but not at T3. Compared to BF, BM at T1 showed a higher representation of Enterococcus, Streptococcus, Enterobacter, Lactococcus, and Propionibacterium. Conclusions: We found a basal state of inflammation in the infants' intestine based on inflammation markers. One month after birth, infants receiving BF exhibited higher levels of inflammation compared to BM.
Collapse
Affiliation(s)
- Juan C Ossa
- Departamento de Pediatría y Cirugía Infantil, Facultad de Medicina, Hospital Dr. Luis Calvo Mackenna, Universidad de Chile, Santiago, Chile
| | - Dominique Yáñez
- Departamento de Pediatría y Cirugía Infantil, Facultad de Medicina, Hospital Dr. Luis Calvo Mackenna, Universidad de Chile, Santiago, Chile
| | - Romina Valenzuela
- Departamento de Pediatría y Cirugía Infantil, Facultad de Medicina, Hospital Dr. Luis Calvo Mackenna, Universidad de Chile, Santiago, Chile
| | - Pablo Gallardo
- Departamento de Pediatría y Cirugía Infantil, Facultad de Medicina, Hospital Dr. Luis Calvo Mackenna, Universidad de Chile, Santiago, Chile
| | - Yalda Lucero
- Departamento de Pediatría y Cirugía Infantil, Facultad de Medicina, Hospital Dr. Luis Calvo Mackenna, Universidad de Chile, Santiago, Chile
| | - Mauricio J Farfán
- Departamento de Pediatría y Cirugía Infantil, Facultad de Medicina, Hospital Dr. Luis Calvo Mackenna, Universidad de Chile, Santiago, Chile
| |
Collapse
|
39
|
Nie Y, Luo F, Lin Q. Dietary nutrition and gut microflora: A promising target for treating diseases. Trends Food Sci Technol 2018. [DOI: 10.1016/j.tifs.2018.03.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
40
|
HIV-exposure, early life feeding practices and delivery mode impacts on faecal bacterial profiles in a South African birth cohort. Sci Rep 2018; 8:5078. [PMID: 29567959 PMCID: PMC5864830 DOI: 10.1038/s41598-018-22244-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 02/16/2018] [Indexed: 02/07/2023] Open
Abstract
There are limited data on meconium and faecal bacterial profiles from African infants and their mothers. We characterized faecal bacterial communities of infants and mothers participating in a South African birth cohort. Stool and meconium specimens were collected from 90 mothers and 107 infants at birth, and from a subset of 72 and 36 infants at 4-12 and 20-28 weeks of age, respectively. HIV-unexposed infants were primarily exclusively breastfed at 4-12 (49%, 26/53) and 20-28 weeks (62%, 16/26). In contrast, HIV-exposed infants were primarily exclusively formula fed at 4-12 (53%; 10/19) and 20-28 weeks (70%, 7/10). Analysis (of the bacterial 16S rRNA gene sequences of the V4 hypervariable region) of the 90 mother-infant pairs showed that meconium bacterial profiles [dominated by Proteobacteria (89%)] were distinct from those of maternal faeces [dominated by Firmicutes (66%) and Actinobacteria (15%)]. Actinobacteria predominated at 4-12 (65%) and 20-28 (50%) weeks. HIV-exposed infants had significantly higher faecal bacterial diversities at both 4-12 (p = 0.026) and 20-28 weeks (p = 0.002). HIV-exposed infants had lower proportions of Bifidobacterium (p = 0.010) at 4-12 weeks. Maternal faecal bacterial profiles were influenced by HIV status, feeding practices and mode of delivery. Further longitudinal studies are required to better understand how these variables influence infant and maternal faecal bacterial composition.
Collapse
|
41
|
Grochowska M, Wojnar M, Radkowski M. The gut microbiota in neuropsychiatric disorders. Acta Neurobiol Exp (Wars) 2018. [DOI: 10.21307/ane-2018-008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
42
|
Mancuso C, Santangelo R. Alzheimer's disease and gut microbiota modifications: The long way between preclinical studies and clinical evidence. Pharmacol Res 2017; 129:329-336. [PMID: 29233677 DOI: 10.1016/j.phrs.2017.12.009] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 12/15/2022]
Abstract
Recent studies have suggested the role of an infectious component in the pathogenesis of Alzheimer's disease (AD). In light of this, research has focused on some bacteria constituting the intestinal microbial flora which can produce amyloid. Once generated, the latter hypothetically triggers a systemic inflammatory response which compromises complex brain functions, such as learning and memory. Clinical studies have shown that, in cognitively impaired elderly patients with brain amyloidosis, there is lower abundance in the gut of E. rectale and B. fragilis, two bacterial species which have an anti-inflammatory activity, versus a greater amount of pro-inflammatory genera such as Escherichia/Shigella. According to these findings, some clinical studies have demonstrated that supplementation with Lactobacilli- and Bifidobacteria- based probiotics has improved cognitive, sensory and emotional functions in subjects with AD. Moreover, certain herbal products, in particular dietetic polyphenols, have proved capable of restoring dysbiosis and, therefore, their prebiotic role could be effective in counteracting the onset of AD regardless of their activity of free radical scavenging or enhancement of the cell stress response. One of the recent greatest novelties in the field of neurodegenerative diseases is the chance to prevent or slow down AD progression with agents, such as probiotics and prebiotics, acting outside the central nervous system.
Collapse
Affiliation(s)
- Cesare Mancuso
- Institute of Pharmacology, Catholic University School of Medicine, Largo F. Vito, 1-00168 Rome, Italy.
| | - Rosaria Santangelo
- Institute of Microbiology, Catholic University School of Medicine, Largo F. Vito, 1-00168 Rome, Italy
| |
Collapse
|
43
|
Milani C, Duranti S, Bottacini F, Casey E, Turroni F, Mahony J, Belzer C, Delgado Palacio S, Arboleya Montes S, Mancabelli L, Lugli GA, Rodriguez JM, Bode L, de Vos W, Gueimonde M, Margolles A, van Sinderen D, Ventura M. The First Microbial Colonizers of the Human Gut: Composition, Activities, and Health Implications of the Infant Gut Microbiota. Microbiol Mol Biol Rev 2017; 81:e00036-17. [PMID: 29118049 PMCID: PMC5706746 DOI: 10.1128/mmbr.00036-17] [Citation(s) in RCA: 1099] [Impact Index Per Article: 137.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The human gut microbiota is engaged in multiple interactions affecting host health during the host's entire life span. Microbes colonize the neonatal gut immediately following birth. The establishment and interactive development of this early gut microbiota are believed to be (at least partially) driven and modulated by specific compounds present in human milk. It has been shown that certain genomes of infant gut commensals, in particular those of bifidobacterial species, are genetically adapted to utilize specific glycans of this human secretory fluid, thus representing a very intriguing example of host-microbe coevolution, where both partners are believed to benefit. In recent years, various metagenomic studies have tried to dissect the composition and functionality of the infant gut microbiome and to explore the distribution across the different ecological niches of the infant gut biogeography of the corresponding microbial consortia, including those corresponding to bacteria and viruses, in healthy and ill subjects. Such analyses have linked certain features of the microbiota/microbiome, such as reduced diversity or aberrant composition, to intestinal illnesses in infants or disease states that are manifested at later stages of life, including asthma, inflammatory bowel disease, and metabolic disorders. Thus, a growing number of studies have reported on how the early human gut microbiota composition/development may affect risk factors related to adult health conditions. This concept has fueled the development of strategies to shape the infant microbiota composition based on various functional food products. In this review, we describe the infant microbiota, the mechanisms that drive its establishment and composition, and how microbial consortia may be molded by natural or artificial interventions. Finally, we discuss the relevance of key microbial players of the infant gut microbiota, in particular bifidobacteria, with respect to their role in health and disease.
Collapse
Affiliation(s)
- Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Sabrina Duranti
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Francesca Bottacini
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Eoghan Casey
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Jennifer Mahony
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Susana Delgado Palacio
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Silvia Arboleya Montes
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Leonardo Mancabelli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Juan Miguel Rodriguez
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Lars Bode
- Department of Pediatrics and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California-San Diego, La Jolla, California, USA
| | - Willem de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- Department of Bacteriology & Immunology, RPU Immunobiology, University of Helsinki, Helsinki, Finland
| | - Miguel Gueimonde
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Abelardo Margolles
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| |
Collapse
|
44
|
Cereal products derived from wheat, sorghum, rice and oats alter the infant gut microbiota in vitro. Sci Rep 2017; 7:14312. [PMID: 29085002 PMCID: PMC5662621 DOI: 10.1038/s41598-017-14707-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 10/12/2017] [Indexed: 12/20/2022] Open
Abstract
The introduction of different nutrient and energy sources during weaning leads to significant changes in the infant gut microbiota. We used an in vitro infant digestive and gut microbiota model system to investigate the effect of four commercially available cereal products based on either wheat, sorghum, rice or oats, on the gut microbiota of six infants. Our results indicated cereal additions induced numerous changes in the gut microbiota composition. The relative abundance of bacterial families associated with fibre degradation, Bacteroidaceae, Bifidobacteriaceae, Lactobacillaceae, Prevotellaceae, Ruminococcaceae and Veillonellaceae increased, whilst the abundance of Enterobacteriaceae decreased with cereal additions. Corresponding changes in the production of SCFAs showed higher concentrations of acetate following all cereal additions, whilst, propionate and butyrate varied between specific cereal additions. These cereal-specific variations in the concentrations of SCFAs showed a moderate correlation with the relative abundance of potential SCFA-producing bacterial families. Overall, our results demonstrated clear shifts in the abundance of bacterial groups associated with weaning and an increase in the production of SCFAs following cereal additions.
Collapse
|
45
|
The microbiota in pediatric rheumatic disease: epiphenomenon or therapeutic target? Curr Opin Rheumatol 2017; 28:537-43. [PMID: 27286235 DOI: 10.1097/bor.0000000000000312] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW There has been increasing interest in the contents and function of the microbiota, as it relates to pediatric inflammatory diseases. Here, we discuss the factors underlying the development of the microbiota, its role in juvenile idiopathic arthritis (JIA) and prospects for therapeutic interventions in the microbiota. RECENT FINDINGS The human microbiota undergoes a succession of changes, until it reaches a mature form. A variety of early-life exposures, including mode of delivery and form of feeding, can affect the contents of the microbiota and possibly impact upon long-term risk of developing autoimmune diseases. The microbiota is altered in children with JIA, including elevated Bacteroides genus in JIA as a whole and decreased Faecalibacterium prausnitzii in pediatric spondyloarthritis. Although there are limited data so far indicating that microbiota-based therapies can result in therapeutic improvement of arthritis, most of the data are on adults and thus may not be applicable to children. SUMMARY Perturbations of the microbiota during childhood may result in the development of a microbiota associated with increased risk of pediatric rheumatic illness. Whether the microbiota can be targeted is a focus of ongoing research.
Collapse
|
46
|
Abstract
Microorganisms in humans form complex communities with important functions and differences in each part of the body. The stomach was considered to be a sterile organ until the discovery of Helicobacter pylori, but nowadays, it is possible to demonstrate that other microorganisms beyond H. pylori can colonize the gastric mucosa and that the diverse microbiota ecosystem of the stomach is different from the mouth and the esophagus, and also from the small intestine and large intestine. H. pylori seems to be the most important member of the gastric microbiota with the highest relative abundance when present, but when it is absent, the stomach has a diverse microbiota. Proteobacteria, Firmicutes, Actinobacteria, Bacteroidetes, and Fusobacteria are the most abundant phyla in both H. pylori-positive and H. pylori-negative patients. The gastric commensal flora may play some role in the H. pylori-associated carcinogenicity, and differences in the gastric microbiota composition of patients with gastric cancer, intestinal metaplasia, and chronic gastritis are described. The gastric microbiota changed gradually from non-atrophic gastritis to intestinal metaplasia, and to gastric cancer (type intestinal).
Collapse
|
47
|
Rutayisire E, Huang K, Liu Y, Tao F. The mode of delivery affects the diversity and colonization pattern of the gut microbiota during the first year of infants' life: a systematic review. BMC Gastroenterol 2016; 16:86. [PMID: 27475754 PMCID: PMC4967522 DOI: 10.1186/s12876-016-0498-0] [Citation(s) in RCA: 382] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 07/21/2016] [Indexed: 12/14/2022] Open
Abstract
Background The human gut is the habitat for diverse and dynamic microbial ecosystem. The human microbiota plays a critical role in functions that sustain health and is a positive asset in host defenses. Establishment of the human intestinal microbiota during infancy may be influenced by multiple factors including delivery mode. Present review compiles existing evidences on the effect of delivery mode on the diversity and colonization pattern of infants gut microbiota. Methods Two investigators searched for relevant scientific publications from four databases (Pubmed, Medline, Embase, and Web of Science). The last search was performed on September 21, 2015, using key terms ((delivery mode OR caesarean delivery OR cesarean section OR vaginal delivery) AND (gut microbiota OR gut microbiome OR gut microflora OR intestinal microflora OR microbial diversity) AND (infants OR children)). All included studies described at least two types of gut microbiota in relation to delivery mode (caesarean section vs vaginal delivery) and used fecal samples to detect gut microbiota. Results Seven out of 652 retrieved studies met inclusion criteria, were included in systematic analysis. Caesarean Section (CS) was associated with both lower abundance and diversity of the phyala Actinobacteria and Bacteroidetes, and higher abundance and diversity of the phylum Firmicute from birth to 3 months of life. At the colonization level, Bifidobacterium, and Bacteroides genera seems to be significantly more frequent in vaginally delivered infants compared with CS delivered. These infants were more colonized by the Clostridium, and Lactobacillus genera. From the reports, it is tempting to say that delivery mode has less effect on colonization and diversity of Bifidobacteria, Bacteroides, Clostridium, and Lactobacillus genera from the age of 6 to 12 months of life. Conclusion The diversity and colonization pattern of the gut microbiota were significantly associated to the mode of delivery during the first three months of life, however the observed significant differences disappears after 6 months of infants life. The healthy gut microbiota is considered to promote development and maturation of the immune system while abnormal gut is considered as the major cause of severe gastrointestinal infections during the infancy. Further studies should investigate the diversity and colonization levels of infant gut microbiota in relation to the mode of delivery and its broad impact on infants’ health at each stage of life.
Collapse
Affiliation(s)
- Erigene Rutayisire
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Meishan Road 81, Hefei City, Anhui Province, 230032, People's Republic of China
| | - Kun Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Meishan Road 81, Hefei City, Anhui Province, 230032, People's Republic of China.,Anhui Provincial Key Laboratory of Population Health & Aristogenics, Hefei City, Anhui Province, People's Republic of China
| | - Yehao Liu
- Department of Public Health Inspection and Quarantine Science, School of Public Health, Anhui Medical University, Hefei City, Anhui Province, People's Republic of China
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Meishan Road 81, Hefei City, Anhui Province, 230032, People's Republic of China. .,Anhui Provincial Key Laboratory of Population Health & Aristogenics, Hefei City, Anhui Province, People's Republic of China.
| |
Collapse
|
48
|
Early-Life Events, Including Mode of Delivery and Type of Feeding, Siblings and Gender, Shape the Developing Gut Microbiota. PLoS One 2016; 11:e0158498. [PMID: 27362264 PMCID: PMC4928817 DOI: 10.1371/journal.pone.0158498] [Citation(s) in RCA: 288] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 06/16/2016] [Indexed: 02/07/2023] Open
Abstract
Colonization of the infant gut is believed to be critically important for a healthy growth as it influences gut maturation, metabolic, immune and brain development in early life. Understanding factors that influence this process is important, since an altered colonization has been associated with a higher risk of diseases later in life. Fecal samples were collected from 108 healthy neonates in the first half year of life. The composition and functionality of the microbiota was characterized by measuring 33 different bacterial taxa by qPCR/RT qPCR, and 8 bacterial metabolites. Information regarding gender, place and mode of birth, presence of siblings or pets; feeding pattern and antibiotic use was collected by using questionnaires. Regression analysis techniques were used to study associations between microbiota parameters and confounding factors over time. Bacterial DNA was detected in most meconium samples, suggesting bacterial exposure occurs in utero. After birth, colonization by species of Bifidobacterium, Lactobacillus and Bacteroides was influenced by mode of delivery, type of feeding and presence of siblings, with differences found at species level and over time. Interestingly, infant-type bifidobacterial species such as B. breve or B. longum subsp infantis were confirmed as early colonizers apparently independent of the factors studied here, while B. animalis subsp. lactis presence was found to be dependent solely on the type of feeding, indicating that it might not be a common infant gut inhabitant. One interesting and rather unexpected confounding factor was gender. This study contributes to our understanding of the composition of the microbiota in early life and the succession process and the evolution of the microbial community as a function of time and events occurring during the first 6 months of life. Our results provide new insights that could be taken into consideration when selecting nutritional supplementation strategies to support the developing infant gut microbiome.
Collapse
|
49
|
Fecal Microbiota and Diet of Children with Chronic Constipation. Int J Pediatr 2016; 2016:6787269. [PMID: 27418934 PMCID: PMC4935906 DOI: 10.1155/2016/6787269] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 05/18/2016] [Indexed: 12/12/2022] Open
Abstract
Many factors explain dysbiosis in chronic constipation (CC), such as a low-fiber diet. The objective of this study was to compare the fecal microbiota of constipated and nonconstipated children and their intake frequencies of food. Methods. This observational study included 79 children (M/F 43/36) aged six to 36 months divided into two groups: cases (39 constipated children) and controls (40 nonconstipated children). We used a structured form to collect demographic variables, conducted anthropometric assessment, and collected food intake frequency data. The fecal microbiota of the stool samples was analyzed by real-time polymerase chain reaction (PCR) using the fluorophore SYBR® Green. Results. Constipated children had a smaller concentration of Lactobacillus per milligram of stool (p = 0.015) than nonconstipated children, but the concentration of Bifidobacterium per milligram of stool (p = 0.323) and the intake of fruits, vegetables (p = 0.563), and junk food (p = 0.093) of the two groups did not differ. Constipated children consumed more dairy products (0.45 ± 0.8; p > 0.001), were more frequently delivered via caesarean section (69.2%), were weaned earlier (median: 120; 60Q1–240Q3), and had a family history of constipation (71.8%). Conclusions. Children with CC have a smaller concentration of Lactobacillus in their stools and consume more dairy products.
Collapse
|
50
|
Austin S, De Castro CA, Bénet T, Hou Y, Sun H, Thakkar SK, Vinyes-Pares G, Zhang Y, Wang P. Temporal Change of the Content of 10 Oligosaccharides in the Milk of Chinese Urban Mothers. Nutrients 2016; 8:E346. [PMID: 27338459 PMCID: PMC4924187 DOI: 10.3390/nu8060346] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/20/2016] [Accepted: 05/31/2016] [Indexed: 12/13/2022] Open
Abstract
Breastfed infants tend to be less prone to infections and may have improved cognitive benefits compared to formula-fed infants. Human milk oligosaccharides (HMO) are the third most abundant component of human milk, but are absent from formulae. They may be partially responsible for the benefits of breastfeeding. In this cross-sectional observational study, the HMO composition of milk from Chinese mothers was studied to determine the impact of stage of lactation, mode of delivery and geographical location. The content of 10 HMO was measured by HPLC in 446 milk samples from mothers living in three different cities in China. Around 21% of the samples contained levels of 2'-fucosyllactose (2'-FL) below the limit of quantification, which is similar to the frequency of fucosyltransferase-2 non-secretors in other populations, but 2'-FL was detected in all samples. Levels of most of the HMO studied decreased during the course of lactation, but the level of 3-fucosyllactose increased. Levels of 2'-FL and 3-fucosyllactose seem to be strongly correlated, suggesting some sort of mechanism for co-regulation. Levels of 6'-sialyllactose were higher than those of 3'-sialyllactose at early stages of lactation, but beyond 2-4 months, 3'-sialyllactose was predominant. Neither mode of delivery nor geographical location had any impact on HMO composition.
Collapse
Affiliation(s)
- Sean Austin
- Nestlé Research Centre, Vers-Chez-Les-Blanc, Lausanne 1000, Switzerland.
| | - Carlos A De Castro
- Nestlé Research Centre, Vers-Chez-Les-Blanc, Lausanne 1000, Switzerland.
| | - Thierry Bénet
- Nestlé Research Centre, Vers-Chez-Les-Blanc, Lausanne 1000, Switzerland.
| | - Yangfeng Hou
- Nestlé Research Center Beijing, Beijing 100095, China.
| | - Henan Sun
- Nestlé Research Center Beijing, Beijing 100095, China.
| | - Sagar K Thakkar
- Nestlé Research Centre, Vers-Chez-Les-Blanc, Lausanne 1000, Switzerland.
| | | | - Yumei Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University Health Science Center, Beijing 100191, China.
| | - Peiyu Wang
- Department of Social Medicine and Health Education, School of Public Health, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|