101
|
Malmuthuge N, Griebel PJ. A Novel Animal Model for Regional Microbial Dysbiosis of the Pioneer Microbial Community. Front Microbiol 2019; 10:1706. [PMID: 31396198 PMCID: PMC6668574 DOI: 10.3389/fmicb.2019.01706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/10/2019] [Indexed: 01/17/2023] Open
Abstract
Pioneer microbiota colonizing the newborn gastrointestinal tract has long-lasting effects on host health. Restoration of the gut microbial community, following dysbiosis during the neonatal period, may be one strategy to prevent undesirable health outcomes linked to an altered neonatal gut microbiome. Without appropriate animal models that recreate the prolonged human neonatal developmental period it is not possible to effectively analyze interventions designed to restore regional microbial populations. Our study used a lamb model in which intestinal segments were surgically isolated (blind-ended) in fetal lambs to create early microbial dysbiosis by delaying post-natal exposure to intestinal ingesta. Intestinal segments isolated in utero retained blood flow, innervation, and lymphatic drainage through the mesenteric attachment. Continuity of the fetal gastro-intestinal tract was re-established by side-to-side anastomosis of intestine proximal and distal to each isolated intestinal segment. Microbial restoration was then implemented in neonatal lambs by reconnecting a portion of the in utero isolated intestinal segments to adjacent intestinal tract 1 and 7 days after birth. Bacterial communities colonizing the adjacent intestine, in utero isolated intestinal segments, and reconnected intestinal segments were profiled using 16S amplicon sequencing on days 1, 7, and 56 of age. The in utero isolated intestinal segments were colonized 1 day after birth but the density of active bacteria was reduced and community composition altered when compared to adjacent intestine. Proteobacteria dominated the adjacent small intestine at early time points (day 1 and day 7) with a shift to primarily Firmicutes on day 56, consistent with establishment of an anaerobic bacterial community. In contrast, Proteobacteria persisted as the predominant community for 56 days in the in utero isolated intestinal segments. There was, however, almost full restoration of the microbial community composition in the in utero isolated intestinal segments following reconnection to the adjacent intestine. The density of beneficial bacteria, especially Bifidobacterium, remained significantly lower in the reconnected intestinal segments at 56 days when compared to adjacent intestine. Post-natal persistence of a stable pioneer community (Proteobacteria) in the in utero isolated intestinal segments provides a model system to study the temporal effects of regional microbial dysbiosis throughout a prolonged neonatal period.
Collapse
Affiliation(s)
- Nilusha Malmuthuge
- Vaccine and Infectious Disease Organization-International Vaccine Centre, University of Saskatchewan, Saskatoon, SK, Canada
| | - Philip J. Griebel
- Vaccine and Infectious Disease Organization-International Vaccine Centre, University of Saskatchewan, Saskatoon, SK, Canada
- School of Public Health, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
102
|
McKeen S, Young W, Fraser K, Roy NC, McNabb WC. Glycan Utilisation and Function in the Microbiome of Weaning Infants. Microorganisms 2019; 7:microorganisms7070190. [PMID: 31277402 PMCID: PMC6681113 DOI: 10.3390/microorganisms7070190] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 06/23/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022] Open
Abstract
Glycans are present exogenously in the diet, expressed and secreted endogenously by host cells, and produced by microbes. All of these processes result in them being available to the gut microbiome, firmly placing glycans at the interface of diet–microbe–host interactions. The most dramatic shift in dietary sources of glycans occurs during the transition from the milk-based neonatal diet to the diverse omnivorous adult diet, and this has profound effects on the composition of the gut microbiome, gene expression by microbes and host cells, mucin composition, and immune development from innate towards adaptive responses. Understanding the glycan-mediated interactions occurring during this transitional window may inform dietary recommendations to support gut and immune development during a vulnerable age. This review aims to summarise the current state of knowledge on dietary glycan mediated changes that may occur in the infant gut microbiome and immune system during weaning.
Collapse
Affiliation(s)
- Starin McKeen
- Food Nutrition & Health, AgResearch, Grasslands Research Centre, Private Bag 11008, Palmerston north 4442, New Zealand
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1023, New Zealand
| | - Wayne Young
- Food Nutrition & Health, AgResearch, Grasslands Research Centre, Private Bag 11008, Palmerston north 4442, New Zealand
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1023, New Zealand
| | - Karl Fraser
- Food Nutrition & Health, AgResearch, Grasslands Research Centre, Private Bag 11008, Palmerston north 4442, New Zealand
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1023, New Zealand
| | - Nicole C Roy
- Food Nutrition & Health, AgResearch, Grasslands Research Centre, Private Bag 11008, Palmerston north 4442, New Zealand
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1023, New Zealand
| | - Warren C McNabb
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland 1023, New Zealand.
| |
Collapse
|
103
|
Grabinger T, Glaus Garzon JF, Hausmann M, Geirnaert A, Lacroix C, Hennet T. Alleviation of Intestinal Inflammation by Oral Supplementation With 2-Fucosyllactose in Mice. Front Microbiol 2019; 10:1385. [PMID: 31275292 PMCID: PMC6593069 DOI: 10.3389/fmicb.2019.01385] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 06/03/2019] [Indexed: 12/30/2022] Open
Abstract
Milk oligosaccharides exert a prebiotic action that contributes to the development of the infant gut microbiota during lactation. Given that milk oligosaccharides remain intact after passage through stomach and small intestine, they can potentially influence the composition of the gut microbiota when ingested as dietary supplements after weaning. To address the regulatory effects of specific oligosaccharides in colitis linked to the microbiota composition, we have supplemented interleukin-10 null (Il10 -/-) mice with four fucosylated and sialylated oligosaccharides. We found that oral supplementation with 2-fucosyllactose significantly decreased the severity of colitis as displayed by reduced inflammatory marker expression, histological and diarrhea scores, an increased epithelial integrity and less pronounced colon shortening. Oral supplementation with 2-fucosyllactose led to a marked expansion of the commensal Ruminococcus gnavus, which was accompanied by an enhanced cecal concentration of propionate. Decreased activation of immune cells by R. gnavus was confirmed by reconstitution of antibiotic-treated Il10 -/- mice and by stimulation of dendritic cells in vitro. This study demonstrates that post-weaning administration of specific oligosaccharides can shift the composition of the gut microbiota to lessen chronic inflammation as observed in Il10 -/- mice. The expansion of R. gnavus sets a positive microbial environment at the cost of pro-inflammatory Gram-negative bacteria, thereby lowering intestinal inflammation.
Collapse
Affiliation(s)
- Thomas Grabinger
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | - Martin Hausmann
- Department of Gastroenterology and Hepatology, University Hospital Zurich - University of Zurich, Zurich, Switzerland
| | - Annelies Geirnaert
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Thierry Hennet
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
104
|
The Effects of Fecal Donors with Different Feeding Patterns on Diarrhea in a Patient Undergoing Hematopoietic Stem Cell Transplantation. Case Rep Hematol 2019; 2019:4505238. [PMID: 31049232 PMCID: PMC6462343 DOI: 10.1155/2019/4505238] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/20/2018] [Accepted: 02/27/2019] [Indexed: 12/14/2022] Open
Abstract
Almost 90% of patients undergoing hematopoietic stem cell transplantation (HSCT) experience diarrheal episodes, which represent a severe, often life-threatening complication for these patients. Although fecal microbiota transplantation (FMT) represents an alternative treatment option for infection-related diarrhea, the application of FMT in HSCT patients is greatly restricted for safety reasons. Furthermore, the therapeutic outcomes of FMT as a diarrhea treatment are somewhat related to the choice of the FMT donor. Here, we comprehensively profiled the dynamic changes in the intestinal microbiota after FMT from two donors with different feeding patterns and the same severely diarrheal recipient undergoing HSCT via a 45-day clinical observation. Importantly, no adverse events attributed to FMT were observed. The stool volume and frequency of the patient were reduced when we used feces from donor #1 (mixed feeding), but these changes were not observed after FMT from donor #2 (exclusive breastfeeding). Interestingly, no obvious differences in overall diversity (Shannon) or richness (Chao1) between the two donors were observed. Additionally, Bifidobacterium accounted for 29.9% and 18.1% of OTUs in the stools of donors #1 and #2, respectively. Lactobacillus accounted for 16.3% and 2.9% of the stools of donors #1 and #2, respectively. Furthermore, through longitudinal monitoring of the patient, we identified 6 OTUs that were particularly sensitive to the different FMT complements. Together, we present a case report suggesting that the overall diversity of the intestinal microbiota may not be the only important element in the selection of an effective FMT donor.
Collapse
|
105
|
Bifidobacterium bifidum ATCC 15696 and Bifidobacterium breve 24b Metabolic Interaction Based on 2'- O-Fucosyl-Lactose Studied in Steady-State Cultures in a Freter-Style Chemostat. Appl Environ Microbiol 2019; 85:AEM.02783-18. [PMID: 30683741 DOI: 10.1128/aem.02783-18] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 01/16/2019] [Indexed: 02/07/2023] Open
Abstract
Infants fed breast milk harbor a gut microbiota in which bifidobacteria are generally predominant. The metabolic interactions of bifidobacterial species need investigation because they may offer insight into the colonization of the gut in early life. Bifidobacterium bifidum ATCC 15696 hydrolyzes 2'-O-fucosyl-lactose (2FL; a major fucosylated human milk oligosaccharide) but does not use fucose released into the culture medium. However, fucose is a growth substrate for Bifidobacterium breve 24b, and both strains utilize lactose for growth. The provision of fucose and lactose by B. bifidum (the donor) allowing the growth of B. breve (the beneficiary) conforms to the concept of syntrophy, but both strains will compete for lactose to multiply. To determine the metabolic impact of this syntrophic/competitive relationship on the donor, the transcriptomes of B. bifidum were determined and compared in steady-state monoculture and coculture using transcriptome sequencing (RNA-seq) and reverse transcription-quantitative PCR (RT-qPCR). B. bifidum genes upregulated in coculture included those encoding alpha-l-fucosidase and carbohydrate transporters and those involved in energy production and conversion. B. bifidum abundance was the same in coculture as in monoculture, but B. breve dominated the coculture numerically. Cocultures during steady-state growth in 2FL medium produced mostly acetate with little lactate (acetate:lactate molar ratio, 8:1) compared to that in monobatch cultures containing lactose (2:1), which reflected the maintenance of steady-state cells in log-phase growth. Darwinian competition is an implicit feature of bacterial communities, but syntrophy is a phenomenon putatively based on cooperation. Our results suggest that the regulation of syntrophy, in addition to competition, may shape bacterial communities.IMPORTANCE This study addresses the microbiology and function of a natural ecosystem (the infant bowel) using in vitro experimentation with bacterial cultures maintained under controlled growth and environmental conditions. We studied the growth of bifidobacteria whose nutrition centered on the hydrolysis of a human milk oligosaccharide. The results revealed responses relating to metabolism occurring in a Bifidobacterium bifidum strain when it provided nutrients that allowed the growth of Bifidobacterium breve, and so discovered biochemical features of these bifidobacteria in relation to metabolic interaction in the shared environment. These kinds of experiments are essential in developing concepts of bifidobacterial ecology that relate to the development of the gut microbiota in early life.
Collapse
|
106
|
Gotoh A, Ojima MN, Katayama T. Minority species influences microbiota formation: the role of Bifidobacterium with extracellular glycosidases in bifidus flora formation in breastfed infant guts. Microb Biotechnol 2019; 12:259-264. [PMID: 30637938 PMCID: PMC6389856 DOI: 10.1111/1751-7915.13366] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 12/14/2018] [Indexed: 12/13/2022] Open
Abstract
The human body houses a variety of microbial ecosystems, such as the microbiotas on the skin, in the oral cavity and in the digestive tract. The gut microbiota is one such ecosystem that contains trillions of bacteria, and it is well established that it can significantly influence host health and diseases. With the advancement in bioinformatics tools, numerous comparative studies based on 16S ribosomal RNA (rRNA) gene sequences, metabolomics, pathological and epidemical analyses have revealed the correlative relationship between the abundance of certain taxa and disease states or amount of certain causative bioactive compounds. However, the 16S rRNA-based taxonomic analyses using next-generation sequencing (NGS) technology essentially detect only the majority species. Although the entire gut microbiome consists of 1013 microbial cells, NGS read counts are given in multiples of 106 , making it difficult to determine the diversity of the entire microbiota. Some recent studies have reported instances where certain minority species play a critical role in creating locally stable conditions for other species by stabilizing the fundamental microbiota, despite their low abundance. These minority species act as 'keystone species', which is a species whose effect on the community is disproportionately large compared to its relative abundance. One of the attributes of keystone species within the gut microbiota is its extensive enzymatic capacity for substrates that are rare or difficult to degrade for other species, such as dietary fibres or host-derived complex glycans, like human milk oligosaccharides (HMOs). In this paper, we propose that more emphasis should be placed on minority taxa and their possible role as keystone species in gut microbiota studies by referring to our recent studies on HMO-mediated microbiota formation in the infant gut.
Collapse
Affiliation(s)
- Aina Gotoh
- Graduate School of BiostudiesKyoto UniversitySakyo‐kuKyoto606‐8502Japan
| | | | - Takane Katayama
- Graduate School of BiostudiesKyoto UniversitySakyo‐kuKyoto606‐8502Japan
| |
Collapse
|
107
|
In vitro immunomodulatory effects of human milk oligosaccharides on murine macrophage RAW264.7 cells. Carbohydr Polym 2019; 207:230-238. [DOI: 10.1016/j.carbpol.2018.11.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 11/09/2018] [Accepted: 11/11/2018] [Indexed: 11/21/2022]
|
108
|
Izumi H, Ehara T, Sugahara H, Matsubara T, Mitsuyama E, Nakazato Y, Tsuda M, Shimizu T, Odamaki T, Xiao JZ, Takeda Y. The Combination of Bifidobacterium breve and Three Prebiotic Oligosaccharides Modifies Gut Immune and Endocrine Functions in Neonatal Mice. J Nutr 2019; 149:344-353. [PMID: 30721975 DOI: 10.1093/jn/nxy248] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/03/2018] [Accepted: 09/04/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Several types of oligosaccharides are used in infant formula to improve the gut microbiota of formula-fed infants. We previously reported that a combination of 3 oligosaccharides (lactulose, raffinose, and galacto-oligosaccharides; LRG) and Bifidobacterium breve effectively increased B. breve numbers, acetate, and the expression of several immune- and gut hormone-related mRNAs in neonatal mice gut. OBJECTIVE We investigated whether changes in neonatal gut microbiota alter gut immune and endocrine development. METHODS We first compared postnatal day (PD) 14 with PD21 in C57BL/6J male mouse pups to identify the physiologic immune and endocrine changes during development. In a separate study, we administered phosphate-buffered saline (control group; CON), B. breve M-16V (M-16V), or M-16V + LRG to male mouse pups from PD6 to PD13, and analyzed the gut microbiota and immune and endocrine parameters on PD14 to evaluate whether M-16V + LRG accelerates gut immune and endocrine development. RESULTS The proportion of regulatory T (Treg) cells in the CD4+ cells of large intestinal lamina propria lymphocytes (LPLs) was significantly increased (63% higher) at PD21 compared with PD14. The serum glucagon-like peptide (GLP)-1 tended to be lower (P = 0.0515) and that of GLP-2 was significantly lower (58% lower) at PD21 than at PD14. M-16V + LRG significantly increased the Treg proportion in large intestinal LPL CD4+ cells (20% and 29% higher compared with CON and M-16V, respectively) at PD14. M-16V + LRG also caused significant changes in expression of large intestinal mRNAs that are consistent with developmental progression, and increased serum concentrations of GLP-1 (207% and 311% higher compared with CON and M-16V, respectively) and GLP-2 (57% and 97% higher compared with CON and M-16V, respectively) at PD14. CONCLUSIONS Neonatal administration of M-16V + LRG alters the gut microbiota and enhances gut immune and endocrine development in suckling mice.
Collapse
Affiliation(s)
| | - Tatsuya Ehara
- Wellness & Nutrition Science Institute, R&D Division
| | - Hirosuke Sugahara
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | | | - Eri Mitsuyama
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | - Yuki Nakazato
- Wellness & Nutrition Science Institute, R&D Division
| | - Muneya Tsuda
- Wellness & Nutrition Science Institute, R&D Division
| | | | - Toshitaka Odamaki
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | - Jin-Zhong Xiao
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | | |
Collapse
|
109
|
Insel R, Knip M. Prospects for primary prevention of type 1 diabetes by restoring a disappearing microbe. Pediatr Diabetes 2018; 19:1400-1406. [PMID: 30136344 DOI: 10.1111/pedi.12756] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/14/2018] [Accepted: 08/14/2018] [Indexed: 12/18/2022] Open
Abstract
Prevention of childhood-onset type 1 diabetes has become more urgent with its marked increased incidence in recent decades in the modern world. Temporally associated with the rising incidence of type 1 diabetes, as well as other autoimmune and allergic diseases in childhood in modern times, is the disappearance of Bifidobacterium and specifically Bifidobacterium longum subsp. infantis (B. infantis) predominance in the intestinal microbiota of breastfed, vaginally-delivered infants. B. infantis efficiently metabolizes human milk oligosaccharides (HMOs) without cross-feeding free sugar monomers to other commensals or pathogens and thereby dominates the intestinal microbiota of breastfed infants. Increased levels of short-chain fatty acids (SCFA), which stimulate both immunoregulation and healthy intestinal and pancreatic β-cell function, are generated by B. infantis. Based on recent observations of the intestinal microbiota in early life in young children who develop type 1 diabetes and demonstration of the robust preventive effects of SCFA in animal models of autoimmune diabetes, we hypothesize that restoring a B. infantis-dominant microbiota early in infancy will prevent islet autoimmunity and childhood-onset type 1 diabetes.
Collapse
Affiliation(s)
| | - Mikael Knip
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland.,Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland.,Folkhälsan Research Center, Helsinki, Finland
| |
Collapse
|
110
|
Milan AM, Hodgkinson AJ, Mitchell SM, Prodhan UK, Prosser CG, Carpenter EA, Fraser K, Cameron-Smith D. Digestive Responses to Fortified Cow or Goat Dairy Drinks: A Randomised Controlled Trial. Nutrients 2018; 10:nu10101492. [PMID: 30322081 PMCID: PMC6213413 DOI: 10.3390/nu10101492] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/02/2018] [Accepted: 10/10/2018] [Indexed: 01/11/2023] Open
Abstract
Fortified milk drinks are predominantly manufactured from bovine (cow) sources. Alternative formulations include those prepared with hydrolysed bovine milk proteins or from alternate bovidae species, such as caprine (goat) milk. Currently, there is little data on protein digestive and metabolic responses following ingestion of fortified milk drinks. To examine the digestive and metabolic responses to commercially-available fortified milks, young adults (n = 15 males: 15 females), in a randomised sequence, ingested isonitrogenous quantities of whole cow-protein (WC), whole goat-protein (WG), or partially-hydrolysed whey cow-protein (HC), commercial fortified milks. Plasma amino acid (AA) and hormonal responses were measured at baseline and again at 5 h after ingestion. Paracetamol recovery, breath hydrogen, and subjective digestive responses were also measured. Postprandial plasma AA was similar between WC and WG, while AA appearance was suppressed with HC. Following HC, there was a negative incremental AUC in plasma branched-chain AAs. Further, HC had delayed gastric emptying, increased transit time, and led to exaggerated insulin and GLP-1 responses, in comparison to whole protein formulas. Overall, WC and WG had similar protein and digestive responses with no differences in digestive comfort. Contrastingly, HC led to delayed gastric emptying, attenuated AA appearance, and a heightened circulating insulin response.
Collapse
Affiliation(s)
- Amber M Milan
- Liggins Institute, University of Auckland, 85 Park Road, Grafton, Private Bag 92019, Auckland 1023, New Zealand.
| | - Alison J Hodgkinson
- Food and Bio-based Products, AgResearch, Private Bag 3123, Hamilton 3240, New Zealand.
| | - Sarah M Mitchell
- Liggins Institute, University of Auckland, 85 Park Road, Grafton, Private Bag 92019, Auckland 1023, New Zealand.
| | - Utpal K Prodhan
- Liggins Institute, University of Auckland, 85 Park Road, Grafton, Private Bag 92019, Auckland 1023, New Zealand.
- Department of Food Technology and Nutritional Science, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh.
| | - Colin G Prosser
- Dairy Goat Co-operative (NZ) Ltd., 18 Gallagher Dr, Melville, Hamilton 3206, New Zealand.
| | - Elizabeth A Carpenter
- Dairy Goat Co-operative (NZ) Ltd., 18 Gallagher Dr, Melville, Hamilton 3206, New Zealand.
| | - Karl Fraser
- AgResearch Grasslands, Private Bag 11008, Palmerston North 4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
| | - David Cameron-Smith
- Liggins Institute, University of Auckland, 85 Park Road, Grafton, Private Bag 92019, Auckland 1023, New Zealand.
- AgResearch Grasslands, Private Bag 11008, Palmerston North 4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
| |
Collapse
|
111
|
Sharing of human milk oligosaccharides degradants within bifidobacterial communities in faecal cultures supplemented with Bifidobacterium bifidum. Sci Rep 2018; 8:13958. [PMID: 30228375 PMCID: PMC6143587 DOI: 10.1038/s41598-018-32080-3] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 09/03/2018] [Indexed: 12/21/2022] Open
Abstract
Gut microbiota of breast-fed infants are generally rich in bifidobacteria. Recent studies show that infant gut-associated bifidobacteria can assimilate human milk oligosaccharides (HMOs) specifically among the gut microbes. Nonetheless, little is known about how bifidobacterial-rich communities are shaped in the gut. Interestingly, HMOs assimilation ability is not related to the dominance of each species. Bifidobacterium longum susbp. longum and Bifidobacterium breve are commonly found as the dominant species in infant stools; however, they show limited HMOs assimilation ability in vitro. In contrast, avid in vitro HMOs consumers, Bifidobacterium bifidum and Bifidobacterium longum subsp. infantis, are less abundant in infant stools. In this study, we observed altruistic behaviour by B. bifidum when incubated in HMOs-containing faecal cultures. Four B. bifidum strains, all of which contained complete sets of HMO-degrading genes, commonly left HMOs degradants unconsumed during in vitro growth. These strains stimulated the growth of other Bifidobacterium species when added to faecal cultures supplemented with HMOs, thereby increasing the prevalence of bifidobacteria in faecal communities. Enhanced HMOs consumption by B. bifidum-supplemented cultures was also observed. We also determined the complete genome sequences of B. bifidum strains JCM7004 and TMC3115. Our results suggest B. bifidum-mediated cross-feeding of HMOs degradants within bifidobacterial communities.
Collapse
|
112
|
Wang Z, Jiang S, Ma C, Huo D, Peng Q, Shao Y, Zhang J. Evaluation of the nutrition and function of cow and goat milk based on intestinal microbiota by metagenomic analysis. Food Funct 2018; 9:2320-2327. [PMID: 29577121 DOI: 10.1039/c7fo01780d] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Milk contains nutrients needed by the body, and the main components of different animal milk vary. Accordingly, we evaluated cow and goat milk's nutrition and their effects on the gut microbiota in mice models using a high-throughput 16S rRNA sequencing technology. The intestinal microbiota of mice changed significantly after the intake of cow and goat milk, and the goat milk had a greater effect on the intestinal microbial community than the cow milk. Bifidobacterium, Allobaculum, Olsenella and Akkermansia grew significantly in both cow and goat milk groups compared with the control group, indicating that milk positively affected their growth. We also found that the citrate cycle (TCA cycle), pyruvate metabolism, and amino sugar and nucleotide sugar metabolism, as well as lipoic acid metabolism, were higher in the goat milk group than in the cow milk group. Association analysis of milk components and their representative intestinal microbiota showed that casein, αs1-casein, and β + κ-casein were positively correlated with Enterococcus and Allobaculum, and negatively correlated with Roseburia. Protein and αs2-casein were positively associated with Akkermansia, Bifidobacterium and Eubacterium.
Collapse
Affiliation(s)
- Zhaoxia Wang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, Inner Mongolia, P. R. China
| | | | | | | | | | | | | |
Collapse
|
113
|
Mediation Analysis as a Means of Identifying Dietary Components That Differentially Affect the Fecal Microbiota of Infants Weaned by Modified Baby-Led and Traditional Approaches. Appl Environ Microbiol 2018; 84:AEM.00914-18. [PMID: 30006390 DOI: 10.1128/aem.00914-18] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/02/2018] [Indexed: 12/13/2022] Open
Abstract
The introduction of "solids" (i.e., complementary foods) to the milk-only diet in early infancy affects the development of the gut microbiota. The aim of this study was to determine whether a "baby-led" approach to complementary feeding that encourages the early introduction of an adult-type diet results in alterations of the gut microbiota composition compared to traditional spoon-feeding. The Baby-Led Introduction to SolidS (BLISS) study randomized 206 infants to BLISS (a modified version of baby-led weaning [BLW], the introduction of solids at 6 months of age, followed by self-feeding of family foods) or control (traditional spoon-feeding of purées) groups. Fecal microbiotas and 3-day weighed-diet records were analyzed for a subset of 74 infants at 7 and 12 months of age. The composition of the microbiota was determined by sequencing of 16S rRNA genes amplified by PCR from bulk DNA extracted from feces. Diet records were used to estimate food and dietary fiber intake. Alpha diversity (number of operational taxonomic units [OTUs]) was significantly lower in BLISS infants at 12 months of age (difference [95% confidence interval {CI}] of 31 OTUs [3.4 to 58.5]; P = 0.028), and while there were no significant differences between control and BLISS infants in relative abundances of Bifidobacteriaceae, Enterobacteriaceae, Veillonellaceae, Bacteroidaceae, Erysipelotrichaceae, Lachnospiraceae, or Ruminococcaceae at 7 or 12 months of age, OTUs representing the genus Roseburia were less prevalent in BLISS microbiotas at 12 months. Mediation models demonstrated that the intake of "fruit and vegetables" and "dietary fiber" explained 29% and 25%, respectively, of the relationship between group (BLISS versus control) and alpha diversity.IMPORTANCE The introduction of solid foods (complementary feeding or weaning) to infants leads to more-complex compositions of microbial communities (microbiota or microbiome) in the gut. In baby-led weaning (BLW), infants are given only finger foods that they can pick up and feed themselves-there is no parental spoon-feeding of puréed baby foods-and infants are encouraged to eat family meals. BLW is a new approach to infant feeding that is increasing in popularity in the United States, New Zealand, the United Kingdom, and Canada. We used mediation modeling, commonly used in health research but not in microbiota studies until now, to identify particular dietary components that affected the development of the infant gut microbiota.
Collapse
|
114
|
Goat and cow milk powder-based diets with or without prebiotics influence gut microbial populations and fermentation products in newly weaned rats. FOOD BIOSCI 2018. [DOI: 10.1016/j.fbio.2018.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
115
|
Bifidobacterium pseudolongum in the Ceca of Rats Fed Hi-Maize Starch Has Characteristics of a Keystone Species in Bifidobacterial Blooms. Appl Environ Microbiol 2018; 84:AEM.00547-18. [PMID: 29802187 DOI: 10.1128/aem.00547-18] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/19/2018] [Indexed: 12/18/2022] Open
Abstract
Starches resistant to mammalian digestion are present in foods and pass to the large bowel, where they may be degraded and fermented by the microbiota. Increases in relative abundances of bifidobacteria (blooms) have been reported in rats whose diet was supplemented with Hi-Maize resistant starch. We determined that the bifidobacterial species present in the rat cecum under these circumstances mostly belonged to Bifidobacterium animalis However, cultures of B. animalis isolated from the rats failed to degrade Hi-Maize starch to any extent. In contrast, Bifidobacterium pseudolongum also detected in the rat microbiota had high starch-degrading ability. Transcriptional comparisons showed increased expression of a type 1 pullulanase, alpha-amylase, and glycogen debranching enzyme by B. pseudolongum when cultured in medium containing Hi-Maize starch. Maltose was released into the culture medium, and B. animalis cultures had shorter doubling times in maltose medium than did B. pseudolongum Thus, B. pseudolongum, which was present at a consistently low abundance in the microbiota, but which has extensive enzymatic capacity to degrade resistant starch, showed the attributes of a keystone species associated with the bifidobacterial bloom.IMPORTANCE This study addresses the microbiology and function of a natural ecosystem (the rat gut) using DNA-based observations and in vitro experimentation. The microbial community of the large bowel of animals, including humans, has been studied extensively through the use of high-throughput DNA sequencing methods and advanced bioinformatics analysis. These studies reveal the compositions and genetic capacities of microbiotas but not the intricacies of how microbial communities function. Our work, combining DNA sequence analysis and laboratory experiments with cultured strains of bacteria, revealed that the increased abundance of bifidobacteria in the rat gut, induced by feeding indigestible starch, involved a species that cannot itself degrade the starch (Bifidobacterium animalis) but cohabits with a species that can (Bifidobacterium pseudolongum). B. pseudolongum has the characteristics of a keystone species in the community because it had low abundance but high ability to perform a critical function, the hydrolysis of resistant starch.
Collapse
|
116
|
Comparative genomics and genotype-phenotype associations in Bifidobacterium breve. Sci Rep 2018; 8:10633. [PMID: 30006593 PMCID: PMC6045613 DOI: 10.1038/s41598-018-28919-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/01/2018] [Indexed: 12/26/2022] Open
Abstract
Bifidobacteria are common members of the gastro-intestinal microbiota of a broad range of animal hosts. Their successful adaptation to this particular niche is linked to their saccharolytic metabolism, which is supported by a wide range of glycosyl hydrolases. In the current study a large-scale gene-trait matching (GTM) effort was performed to explore glycan degradation capabilities in B. breve. By correlating the presence/absence of genes and associated genomic clusters with growth/no-growth patterns across a dataset of 20 Bifidobacterium breve strains and nearly 80 different potential growth substrates, we not only validated the approach for a number of previously characterized carbohydrate utilization clusters, but we were also able to discover novel genetic clusters linked to the metabolism of salicin and sucrose. Using GTM, genetic associations were also established for antibiotic resistance and exopolysaccharide production, thereby identifying (novel) bifidobacterial antibiotic resistance markers and showing that the GTM approach is applicable to a variety of phenotypes. Overall, the GTM findings clearly expand our knowledge on members of the B. breve species, in particular how their variable genetic features can be linked to specific phenotypes.
Collapse
|
117
|
tuf Gene Sequence Variation in Bifidobacterium longum subsp. infantis Detected in the Fecal Microbiota of Chinese Infants. Appl Environ Microbiol 2018; 84:AEM.00336-18. [PMID: 29703739 DOI: 10.1128/aem.00336-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 04/20/2018] [Indexed: 12/12/2022] Open
Abstract
Members of the bacterial genus Bifidobacterium generally dominate the fecal microbiota of infants. The species Bifidobacterium longum is prevalent, but the B. longum subsp. longum and B. longum subsp. infantis strains that are known to colonize the infant bowel are not usually differentiated in microbiota investigations. These subspecies differ in their capacities to metabolize human milk oligosaccharides (HMO) and may have different ecological and symbiotic roles in humans. Quantitative PCR provides a quick analytical method by which to accurately ascertain the abundances of target species in microbiotas and microcosms. However, amplification targets in DNA extracted from samples need to be dependably differential. We evaluated the tuf gene sequence as a molecular target for quantitative PCR measurements of the abundances of B. longum subsp. infantis and B. longum subsp. longum in fecal microbiotas. This approach resulted in the detection of a tuf gene variant (operational taxonomic unit 49 [OTU49]) in Chinese infants that has sequence similarities to both B. longum subsp. infantis and B. longum subsp. longum We compared the genome sequence and growth and transcriptional characteristics of an OTU49 isolate cultured in HMO medium to those of other B. longum subsp. infantis cultures. We concluded from these studies that OTU49 belongs to B. longum subsp. infantis, that dependable quantitative PCR (qPCR) differentiation between the B. longum subspecies cannot be achieved by targeting tuf gene sequences, and that functional genes involved in carbohydrate metabolism might be better targets because they delineate ecological functions.IMPORTANCE High-throughput DNA sequencing methods and advanced bioinformatics analysis have revealed the composition and biochemical capacities of microbial communities (microbiota and microbiome), including those that inhabit the gut of human infants. However, the microbiology and function of natural ecosystems have received little attention in recent decades, so an appreciation of the dynamics of gut microbiota interactions is lacking. With respect to infants, rapid methodologies, such as quantitative PCR, are needed to determine the prevalences and proportions of different bifidobacterial species in observational and microcosm studies in order to obtain a better understanding of the dynamics of bifidobacterial nutrition and syntrophy, knowledge that might be used to manipulate the microbiota and perhaps ensure the better health of infants.
Collapse
|
118
|
Denning NL, Prince JM. Neonatal intestinal dysbiosis in necrotizing enterocolitis. Mol Med 2018; 24:4. [PMID: 30134786 PMCID: PMC6016883 DOI: 10.1186/s10020-018-0002-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 02/13/2018] [Indexed: 12/11/2022] Open
Abstract
Necrotizing Enterocolitis (NEC) is one of the most devastating gastrointestinal diseases in neonates, particularly among preterm infants in whom surgical NEC is the leading cause of morbidity. NEC pathophysiology occurs in the hyper-reactive milieu of the premature gut after bacterial colonization. The resultant activation of the TLR4 pathway appears to be a strongly contributing factor. Advancements in metagenomics may yield new clarity to the relationship between the neonatal intestinal microbiome and the development of NEC. After a century without effective directed treatments, microbiome manipulation offers a promising therapeutic target for the prevention and treatment of this devastating disease.
Collapse
Affiliation(s)
- Naomi-Liza Denning
- Division of Pediatric Surgery, Zucker School of Medicine at Hofstra/Northwell, Cohen Children's Medical Center, 269-01 76th Avenue, CH 158, New Hyde Park, New York, NY, 11040, USA. .,Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA.
| | - Jose M Prince
- Division of Pediatric Surgery, Zucker School of Medicine at Hofstra/Northwell, Cohen Children's Medical Center, 269-01 76th Avenue, CH 158, New Hyde Park, New York, NY, 11040, USA.,Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA.,Trauma Institute, Northwell Health System, Manhasset, NY, 11030, USA
| |
Collapse
|
119
|
Molès JP, Tuaillon E, Kankasa C, Bedin AS, Nagot N, Marchant A, McDermid JM, Van de Perre P. Breastmilk cell trafficking induces microchimerism-mediated immune system maturation in the infant. Pediatr Allergy Immunol 2018; 29:133-143. [PMID: 29197124 DOI: 10.1111/pai.12841] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/28/2017] [Indexed: 12/31/2022]
Abstract
Initiating breastfeeding within the first hour of life confers an important benefit in terms of child mortality and severe morbidity. Intestinal permeability to ingested macromolecules and immunoglobulins is limited to the first days of human life. These exchanges cease in the very early post-partum period but may increase beyond the neonatal period in response to local inflammation or introduction of a weaning food. From animal- and limited human-based observations, compelling evidence points out to breastmilk cells also trafficking from mother to infant mucosal tissues and participating to the maternal microchimerism. The precise nature of breastmilk cells that are involved is presently not known but likely includes progenitor/stem cells-representing up to 6% of breastmilk cells-with possible contribution of mature immune cells. Stem cell microchimerism may induce tolerance to non-inherited maternal antigens (NIMAs), breastfeeding generating regulatory T cells (Treg ) that suppress antimaternal immunity. Therefore, in complement to pregnancy-induced microchimerism, breastfeeding-induced microchimerism may be pivotal in infant immune development, intestinal tissue repair/growth and protection against infectious diseases. As a continuum of the gestational period, the neonatal gut may be considered as a temporary, but important developmental extension of the role played by the placenta during intrauterine life; breastmilk playing the role of maternal blood by delivering maternal soluble factors (macromolecules, Ig, cytokines) and immunologically active milk cells. A better understanding of breastfeeding-induced maternal microchimerism would provide further evidence in support of public health messages that reinforce the importance of early initiation of breastfeeding.
Collapse
Affiliation(s)
- Jean-Pierre Molès
- Pathogenesis and Control of Chronic Infections, INSERM, EFS, Université de Montpellier, Montpellier, France
| | - Edouard Tuaillon
- Pathogenesis and Control of Chronic Infections, INSERM, EFS, Université de Montpellier, Montpellier, France.,Department of Bacteriology-Virology and Department of Medical Information, CHU Montpellier, Montpellier, France
| | - Chipepo Kankasa
- Department of Paediatrics and Child Health, School of Medicine, University Teaching Hospital, University of Zambia, Lusaka, Zambia
| | - Anne-Sophie Bedin
- Pathogenesis and Control of Chronic Infections, INSERM, EFS, Université de Montpellier, Montpellier, France
| | - Nicolas Nagot
- Pathogenesis and Control of Chronic Infections, INSERM, EFS, Université de Montpellier, Montpellier, France.,Department of Bacteriology-Virology and Department of Medical Information, CHU Montpellier, Montpellier, France
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles, Brussels, Belgium
| | - Joann M McDermid
- Division of Infectious Diseases & International Health, Department of Medicine, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Philippe Van de Perre
- Pathogenesis and Control of Chronic Infections, INSERM, EFS, Université de Montpellier, Montpellier, France.,Department of Bacteriology-Virology and Department of Medical Information, CHU Montpellier, Montpellier, France
| |
Collapse
|
120
|
Voss JD, Goodson MS, Leon JC. Phenotype diffusion and one health: A proposed framework for investigating the plurality of obesity epidemics across many species. Zoonoses Public Health 2018; 65:279-290. [PMID: 29430857 DOI: 10.1111/zph.12445] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Indexed: 01/07/2023]
Abstract
We propose the idea of "phenotype diffusion," which is a rapid convergence of an observed trait in some human and animal populations. The words phenotype and diffusion both imply observations independent of mechanism as phenotypes are observed traits with multiple possible genetic mechanisms and diffusion is an observed state of being widely distributed. Recognizing shared changes in phenotype in multiple species does not by itself reveal a particular mechanism such as a shared exposure, shared adaptive need, particular stochastic process or a transmission pathway. Instead, identifying phenotype diffusion suggests the mechanism should be explored to help illuminate the ways human and animal health are connected and new opportunities for optimizing these links. Using the plurality of obesity epidemics across multiple species as a prototype for shared changes in phenotype, the goal of this review was to explore eco-evolutionary theories that could inform further investigation. First, evolutionary changes described by hologenome evolution, pawnobe evolution, transposable element (TE) thrust and the drifty gene hypothesis will be discussed within the context of the selection asymmetries among human and animal populations. Secondly, the ecology of common source exposures (bovine milk, xenohormesis and "obesogens"), niche evolution and the hygiene hypothesis will be summarized. Finally, we synthesize these considerations. For example, many agricultural breeds have been aggressively selected for weight gain, microbiota (e.g., adenovirus 36, toxoplasmosis) associated with (or infecting) these breeds cause experimental weight gain in other animals, and these same microbes are associated with human obesity. We propose applications of phenotype diffusion could include zoonotic biosurveillance, biocontainment, antibiotic stewardship and environmental priorities. The One Health field is focused on the connections between the health of humans, animals and the environment, and so identification of phenotype diffusion is highly relevant for practitioners (public health officials, physicians and veterinarians) in this field.
Collapse
Affiliation(s)
- J D Voss
- Epidemiology Consult Service Division, United States Air Force School of Aerospace Medicine, Wright-Patterson AFB, OH, USA
| | - M S Goodson
- 711th Human Performance Wing, Human Effectiveness Directorate, Wright-Patterson AFB, OH, USA.,UES Inc., Dayton, OH, USA
| | - J C Leon
- Epidemiology Consult Service Division, United States Air Force School of Aerospace Medicine, Wright-Patterson AFB, OH, USA
| |
Collapse
|
121
|
The Distal Gut Bacterial Community of Some Primates and Carnivora. Curr Microbiol 2017; 75:213-222. [DOI: 10.1007/s00284-017-1368-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 06/28/2017] [Indexed: 10/18/2022]
|
122
|
Bazanella M, Maier TV, Clavel T, Lagkouvardos I, Lucio M, Maldonado-Gòmez MX, Autran C, Walter J, Bode L, Schmitt-Kopplin P, Haller D. Randomized controlled trial on the impact of early-life intervention with bifidobacteria on the healthy infant fecal microbiota and metabolome. Am J Clin Nutr 2017; 106:1274-1286. [PMID: 28877893 DOI: 10.3945/ajcn.117.157529] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 08/08/2017] [Indexed: 11/14/2022] Open
Abstract
Background: Early-life colonization of the intestinal tract is a dynamic process influenced by numerous factors. The impact of probiotic-supplemented infant formula on the composition and function of the infant gut microbiota is not well defined.Objective: We sought to determine the effects of a bifidobacteria-containing formula on the healthy human intestinal microbiome during the first year of life.Design: A double-blind, randomized, placebo-controlled study of newborn infants assigned to a standard whey-based formula containing a total of 107 colony-forming units (CFU)/g of Bifidobacterium bifidum, Bifidobacterium breve, Bifidobacterium longum, B. longum subspecies infantis (intervention), or to a control formula without bifidobacteria (placebo). Breastfed controls were included. Diversity and composition of fecal microbiota were determined by 16S ribosomal RNA gene amplicon sequencing, and metabolite profiles were analyzed by ultrahigh-performance liquid chromatography-mass spectrometry over a period of 2 y.Results: Infants (n = 106) were randomly assigned to either the interventional (n = 48) or placebo (n = 49) group; 9 infants were exclusively breastfed throughout the entire intervention period of 12 mo. Infants exposed to bifidobacteria-supplemented formula showed decreased occurrence of Bacteroides and Blautia spp. associated with changes in lipids and unknown metabolites at month 1. Microbiota and metabolite profiles of intervention and placebo groups converged during the study period, and long-term colonization (24 mo) of the supplemented Bifidobacterium strains was not detected. Significant differences in microbiota and metabolites were detected between infants fed breast milk and those fed formula (P < 0.005) and between infants birthed vaginally and those birthed by cesarean delivery (P < 0.005). No significant differences were observed between infant feeding groups regarding growth, antibiotic uptake, or other health variables (P > 0.05).Conclusion: The supplementation of bifidobacteria to infant diet can modulate the occurrence of specific bacteria and metabolites during early life with no detectable long-term effects. This trial was registered at germanctr.de as DRKS00003660.
Collapse
Affiliation(s)
| | - Tanja V Maier
- Research Unit Analytical Biogeochemistry, Helmholtz Center Munich, Oberschleißheim, Germany
| | | | | | - Marianna Lucio
- Research Unit Analytical Biogeochemistry, Helmholtz Center Munich, Oberschleißheim, Germany
| | | | - Chloe Autran
- Divisions of Neonatology and Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MoMICoRE), University of California, San Diego, La Jolla, CA
| | - Jens Walter
- Chair for Nutrition, Microbes and Gastrointestinal Health, University of Alberta, Edmonton, Alberta, Canada; and
| | - Lars Bode
- Divisions of Neonatology and Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MoMICoRE), University of California, San Diego, La Jolla, CA
| | - Philippe Schmitt-Kopplin
- Chair of Analytical Food Chemistry, Technical University of Munich, Freising, Germany.,Research Unit Analytical Biogeochemistry, Helmholtz Center Munich, Oberschleißheim, Germany
| | - Dirk Haller
- Chair of Nutrition and Immunology, .,ZIEL - Institute for Food & Health, and
| |
Collapse
|
123
|
Bernstein CN. Review article: changes in the epidemiology of inflammatory bowel disease-clues for aetiology. Aliment Pharmacol Ther 2017; 46:911-919. [PMID: 28949021 DOI: 10.1111/apt.14338] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/06/2017] [Accepted: 09/04/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND The changing epidemiology of inflammatory bowel disease (IBD) in both the developed and developing worlds may provide insights into disease aetiology. Factors that impact on the gut microbiome are leading aetiological candidates. AIM To review epidemiological studies and trends that identify risk factors for the development of IBD. METHODS Studies that identified factors associated with the development of IBD differentially in children and adults were reviewed. There was a focus on epidemiological studies and on studies that involve the gut microbiome. RESULTS Use of antibiotics has been shown to be associated with development of Crohn's disease in childhood (odds ratio, OR = 2.75, 95% CI 1.72-4.38). Breastfeeding has been protective against developing IBD (OR=0.69, 95% CI 0.51-0.94), but there is a paucity of data exploring duration of breastfeeding and timing of introduction of bottled milk or table food. Antibiotics and diet changes can also impact on adults enhancing the risk for IBD. Both smoking (OR=1.76, 95% CI 1.40-2.22) and oral contraceptives (relative risk=1.46, 95% CI 1.26-1.70) increase the risk for Crohn's disease and their use is associated with worse outcomes in Crohn's disease. It is unclear if their impact is mediated through the gut microbiome. CONCLUSIONS A leading aetiological clue for IBD based on epidemiological studies is the antecedent use of antibiotics both for children and adults. Some dietary changes may be a risk for adults but there is a paucity of dietary data in children prior to IBD development. Both antibiotic use and dietary changes have the potential to impact the gut microbiome, which in turn can alter the gut immune response.
Collapse
Affiliation(s)
- C N Bernstein
- University of Manitoba IBD Clinical and Research Centre, Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
124
|
Wong CB, Sugahara H, Odamaki T, Xiao JZ. Different physiological properties of human-residential and non-human-residential bifidobacteria in human health. Benef Microbes 2017; 9:111-122. [PMID: 28969444 DOI: 10.3920/bm2017.0031] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Bifidobacteria have increasingly been shown to exert positive health benefits to humans, which are clearly reflected by their application in various commercialised dairy products and supplements. Bifidobacteria naturally inhabit a range of ecological niches and display substantial differences in their ecological adaptation among species. In general, bifidobacteria could be categorised into two major groups; bifidobacterial species of human origins as human-residential bifidobacteria (HRB) while other species which are the natural inhabitants of animals or environment as non-HRB. Current research has focused on the differential physiological features of HRB and non-HRB, such as metabolic capabilities, whilst comparative and functional genomic investigations have revealed the genetic attributes of bifidobacteria that may explain their colonisation affinities in human gut. It is becoming more apparent that distinct residential origins of bifidobacteria are likely contributed to their comparable adaptive health attributes on human host. Notably, debate still remains about the nature of bifidobacteria for use as human probiotics. Clinical evaluations involving supplementation of bifidobacteria of different origins point out the superiority of HRB in human host. Evidence also suggests that HRB especially infant-type HRB may exert better health-promoting effects and therefore serve as a better probiotic candidate for infant use. In this review, we aim to provide an overview of the genotypic and physiological differences of bifidobacteria associated with different residential origins and to shed light on the practical considerations for selection of bifidobacteria as probiotics in order to establish a healthy gut microbial community in humans.
Collapse
Affiliation(s)
- C B Wong
- 1 Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama, Kanagawa 252-8583, Japan
| | - H Sugahara
- 1 Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama, Kanagawa 252-8583, Japan
| | - T Odamaki
- 1 Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama, Kanagawa 252-8583, Japan
| | - J Z Xiao
- 1 Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama, Kanagawa 252-8583, Japan
| |
Collapse
|
125
|
Nagpal R, Kurakawa T, Tsuji H, Takahashi T, Kawashima K, Nagata S, Nomoto K, Yamashiro Y. Evolution of gut Bifidobacterium population in healthy Japanese infants over the first three years of life: a quantitative assessment. Sci Rep 2017; 7:10097. [PMID: 28855672 PMCID: PMC5577255 DOI: 10.1038/s41598-017-10711-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 08/15/2017] [Indexed: 01/05/2023] Open
Abstract
Bifidobacteria are important members of human gut microbiota; however, quantitative data on their early-life dynamics is limited. Here, using a sensitive reverse transcription-qPCR approach, we demonstrate the carriage of eight signature infant-associated Bifidobacterium species (B. longum, B. breve, B. bifidum, B. catenulatum group, B. infantis, B. adolescentis, B. angulatum and B. dentium) in 76 healthy full-term vaginally-born infants from first day to three years of life. About 21% babies carry bifidobacteria at first day of life (6.2 ± 1.9 log10 cells/g feces); and this carriage increases to 64% (8.0 ± 2.2), 79% (8.5 ± 2.1), 97% (9.3 ± 1.8), 99% (9.6 ± 1.6), and 100% (9.7 ± 0.9) at age 7 days, 1, 3 and 6 months, and 3 years, respectively. B. longum, B. breve, B. catenulatum group and B. bifidum are among the earliest and abundant bifidobacterial clades. Interestingly, infants starting formula-feed as early as first week of life have higher bifidobacterial carriage compared to exclusively breast-fed counterparts. Bifidobacteria demonstrate an antagonistic correlation with enterobacteria and enterococci. Further analyses also reveal a relatively lower/ delayed bifidobacterial carriage in cesarean-born babies. The study presents a quantitative perspective of the early-life gut Bifidobacterium colonization and shows how factors such as birth and feeding modes could influence this acquisition even in healthy infants.
Collapse
Affiliation(s)
- Ravinder Nagpal
- Probiotics Research Laboratory, Juntendo University Graduate School of Medicine, Hongo 2-9-8-3F, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Gut Microbiome and Metabolic Diseases, Center for Diabetes, Obesity and Metabolism, Wake Forest School of Medicine, Biotech Place, Winston-Salem, NC, 27101, USA.
| | | | | | | | | | - Satoru Nagata
- Department of Pediatrics, School of Medicine, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Koji Nomoto
- Yakult Central Institute, Kunitachi-shi, Tokyo, Japan
| | - Yuichiro Yamashiro
- Probiotics Research Laboratory, Juntendo University Graduate School of Medicine, Hongo 2-9-8-3F, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
126
|
Kato K, Odamaki T, Mitsuyama E, Sugahara H, Xiao JZ, Osawa R. Age-Related Changes in the Composition of Gut Bifidobacterium Species. Curr Microbiol 2017; 74:987-995. [PMID: 28593350 PMCID: PMC5486783 DOI: 10.1007/s00284-017-1272-4] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 05/27/2017] [Indexed: 12/18/2022]
Abstract
Bifidobacteria are one of the major components in human microbiota that are suggested to function in maintaining human health. The colonization and cell number of Bifidobacterium species in human intestine vary with ageing. However, sequential changes of Bifidobacterium species ranging from newborns to centenarians remain unresolved. Here, we investigated the gut compositional changes of Bifidobacterium species over a wide range of ages. Faecal samples of 441 healthy Japanese subjects between the ages of 0 and 104 years were analysed using real-time PCR with species-specific primers. B. longum group was widely detected from newborns to centenarians, with the highest detection rate. B. breve was detected in approximately 70% of children under 3 years old. B. adolescentis and B. catenulatum groups were predominant after weaning. B. bifidum was detected at almost all ages. The detection rate of B. dentium was higher in the elderly than in other ages. B. animalis ssp. lactis was detected in 11.4% of the subjects and their ages were restricted. B. gallinarum goup was detected in only nine subjects, while B. minimum and B. mongoliense were undetected at any age. The presence of certain Bifidobacterium groups was associated with significantly higher numbers of other Bifidobacterium species/subspecies. Inter-species correlations were found among each species, exception for B. animalis ssp. lactis. These results revealed the patterns and transition points with respect to compositional changes of Bifidobacterium species that occur with ageing, and the findings indicate that there may be symbiotic associations between some of these species in the gut microbiota.
Collapse
Affiliation(s)
- Kumiko Kato
- Next Generation Science Institute R&D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama-city, Kanagawa, 252-8583, Japan.
| | - Toshitaka Odamaki
- Next Generation Science Institute R&D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama-city, Kanagawa, 252-8583, Japan
| | - Eri Mitsuyama
- Next Generation Science Institute R&D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama-city, Kanagawa, 252-8583, Japan
| | - Hirosuke Sugahara
- Next Generation Science Institute R&D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama-city, Kanagawa, 252-8583, Japan
| | - Jin-Zhong Xiao
- Next Generation Science Institute R&D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama-city, Kanagawa, 252-8583, Japan
| | - Ro Osawa
- Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, Japan
| |
Collapse
|
127
|
Lawley B, Munro K, Hughes A, Hodgkinson AJ, Prosser CG, Lowry D, Zhou SJ, Makrides M, Gibson RA, Lay C, Chew C, Lee PS, Wong KH, Tannock GW. Differentiation of Bifidobacterium longum subspecies longum and infantis by quantitative PCR using functional gene targets. PeerJ 2017; 5:e3375. [PMID: 28560114 PMCID: PMC5446769 DOI: 10.7717/peerj.3375] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/04/2017] [Indexed: 01/16/2023] Open
Abstract
Background Members of the genus Bifidobacterium are abundant in the feces of babies during the exclusively-milk-diet period of life. Bifidobacterium longum is reported to be a common member of the infant fecal microbiota. However, B. longum is composed of three subspecies, two of which are represented in the bowel microbiota (B. longum subsp. longum; B. longum subsp. infantis). B. longum subspecies are not differentiated in many studies, so that their prevalence and relative abundances are not accurately known. This may largely be due to difficulty in assigning subspecies identity using DNA sequences of 16S rRNA or tuf genes that are commonly used in bacterial taxonomy. Methods We developed a qPCR method targeting the sialidase gene (subsp. infantis) and sugar kinase gene (subsp. longum) to differentiate the subspecies using specific primers and probes. Specificity of the primers/probes was tested by in silico, pangenomic search, and using DNA from standard cultures of bifidobacterial species. The utility of the method was further examined using DNA from feces that had been collected from infants inhabiting various geographical regions. Results A pangenomic search of the NCBI genomic database showed that the PCR primers/probes targeted only the respective genes of the two subspecies. The primers/probes showed total specificity when tested against DNA extracted from the gold standard strains (type cultures) of bifidobacterial species detected in infant feces. Use of the qPCR method with DNA extracted from the feces of infants of different ages, delivery method and nutrition, showed that subsp. infantis was detectable (0–32.4% prevalence) in the feces of Australian (n = 90), South-East Asian (n = 24), and Chinese babies (n = 91), but in all cases at low abundance (<0.01–4.6%) compared to subsp. longum (0.1–33.7% abundance; 21.4–100% prevalence). Discussion Our qPCR method differentiates B. longum subspecies longum and infantis using characteristic functional genes. It can be used as an identification aid for isolates of bifidobacteria, as well as in determining prevalence and abundance of the subspecies in feces. The method should thus be useful in ecological studies of the infant gut microbiota during early life where an understanding of the ecology of bifidobacterial species may be important in developing interventions to promote infant health.
Collapse
Affiliation(s)
- Blair Lawley
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Karen Munro
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Alan Hughes
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | | | | | - Dianne Lowry
- Dairy Goat Cooperative (NZ) Ltd., Hamilton, New Zealand
| | - Shao J Zhou
- Women's and Children's Health Research Institute, Adelaide, Australia.,School of Agriculture, Food and Wine, University of Adelaide, Adelaide, Australia
| | - Maria Makrides
- Healthy Mothers, Babies and Children, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Robert A Gibson
- School of Agriculture, Food and Wine, University of Adelaide, Adelaide, Australia
| | | | | | | | | | - Gerald W Tannock
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.,Riddet Institute Centre of Research Excellence, Palmerston North, New Zealand.,Microbiome Otago, University of Otago, Dunedin, New Zealand
| |
Collapse
|
128
|
Molecular Insight into Evolution of Symbiosis between Breast-Fed Infants and a Member of the Human Gut Microbiome Bifidobacterium longum. Cell Chem Biol 2017; 24:515-524.e5. [PMID: 28392148 DOI: 10.1016/j.chembiol.2017.03.012] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 02/13/2017] [Accepted: 03/13/2017] [Indexed: 12/23/2022]
Abstract
Breast-fed infants generally have a bifidobacteria-rich microbiota with recent studies indicating that human milk oligosaccharides (HMOs) selectively promote bifidobacterial growth. Bifidobacterium bifidum possesses a glycoside hydrolase family 20 lacto-N-biosidase for liberating lacto-N-biose I from lacto-N-tetraose, an abundant HMO unique to human milk, while Bifidobacterium longum subsp. longum has a non-classified enzyme (LnbX). Here, we determined the crystal structure of the catalytic domain of LnbX and provide evidence for creation of a novel glycoside hydrolase family, GH136. The structure, in combination with inhibition and mutation studies, provides insight into the molecular mechanism and broader substrate specificity of this enzyme. Moreover, through genetic studies, we show that lnbX is indispensable for B. longum growth on lacto-N-tetraose and is a key genetic factor for persistence in the gut of breast-fed infants. Overall, this study reveals possible evolutionary routes for the emergence of symbiosis between humans and bifidobacterial species in the infant gut.
Collapse
|
129
|
Tun HM, Konya T, Takaro TK, Brook JR, Chari R, Field CJ, Guttman DS, Becker AB, Mandhane PJ, Turvey SE, Subbarao P, Sears MR, Scott JA, Kozyrskyj AL. Exposure to household furry pets influences the gut microbiota of infant at 3-4 months following various birth scenarios. MICROBIOME 2017; 5:40. [PMID: 28381231 PMCID: PMC5382463 DOI: 10.1186/s40168-017-0254-x] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/14/2017] [Indexed: 05/25/2023]
Abstract
BACKGROUND Early-life exposure to household pets has the capacity to reduce risk for overweight and allergic disease, especially following caesarean delivery. Since there is some evidence that pets also alter the gut microbial composition of infants, changes to the gut microbiome are putative pathways by which pet exposure can reduce these risks to health. To investigate the impact of pre- and postnatal pet exposure on infant gut microbiota following various birth scenarios, this study employed a large subsample of 746 infants from the Canadian Healthy Infant Longitudinal Development Study (CHILD) cohort, whose mothers were enrolled during pregnancy between 2009 and 2012. Participating mothers were asked to report on household pet ownership at recruitment during the second or third trimester and 3 months postpartum. Infant gut microbiota were profiled with 16S rRNA sequencing from faecal samples collected at the mean age of 3.3 months. Two categories of pet exposure (i) only during pregnancy and (ii) pre- and postnatally were compared to no pet exposure under different birth scenarios. RESULTS Over half of studied infants were exposed to at least one furry pet in the prenatal and/or postnatal periods, of which 8% were exposed in pregnancy alone and 46.8% had exposure during both time periods. As a common effect in all birth scenarios, pre- and postnatal pet exposure enriched the abundance of Oscillospira and/or Ruminococcus (P < 0.05) with more than a twofold greater likelihood of high abundance. Among vaginally born infants with maternal intrapartum antibiotic prophylaxis exposure, Streptococcaceae were substantially and significantly reduced by pet exposure (P < 0.001, FDRp = 0.03), reflecting an 80% decreased likelihood of high abundance (OR 0.20, 95%CI, 0.06-0.70) for pet exposure during pregnancy alone and a 69% reduced likelihood (OR 0.31, 95%CI, 0.16-0.58) for exposure in the pre- and postnatal time periods. All of these associations were independent of maternal asthma/allergy status, siblingship, breastfeeding exclusivity and other home characteristics. CONCLUSIONS The impact of pet ownership varies under different birth scenarios; however, in common, exposure to pets increased the abundance of two bacteria, Ruminococcus and Oscillospira, which have been negatively associated with childhood atopy and obesity.
Collapse
Affiliation(s)
- Hein M Tun
- Department of Pediatrics, University of Alberta, 3-527 Edmonton Clinic Health Academy, 11405-87th Avenue, Edmonton, AB, T6G IC9, Canada
| | - Theodore Konya
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Tim K Takaro
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Jeffrey R Brook
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Radha Chari
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - David S Guttman
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, ON, Canada
| | - Allan B Becker
- Department of Pediatrics and Child Health, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Piush J Mandhane
- Department of Pediatrics, University of Alberta, 3-527 Edmonton Clinic Health Academy, 11405-87th Avenue, Edmonton, AB, T6G IC9, Canada
| | - Stuart E Turvey
- Department of Pediatrics, Child & Family Research Institute, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Padmaja Subbarao
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Malcolm R Sears
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - James A Scott
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Anita L Kozyrskyj
- Department of Pediatrics, University of Alberta, 3-527 Edmonton Clinic Health Academy, 11405-87th Avenue, Edmonton, AB, T6G IC9, Canada.
| |
Collapse
|
130
|
Li SS, Miao JJ, Wu ZX, Yao JR, Li M, Yu Q, He F. [A preliminary analysis of changes in composition of intestinal microbiota during infancy using polymerase chain reaction-denaturing gradient gel electrophoresis]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:331-336. [PMID: 28302207 PMCID: PMC7390155 DOI: 10.7499/j.issn.1008-8830.2017.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 01/19/2017] [Indexed: 06/06/2023]
Abstract
OBJECTIVE To investigate the composition of bacteria in the stools of infants and the colonization of intestinal microbiota during infancy. METHODS Fresh stools were collected from 15 healthy infants at 0, 2, 4, 7, 10, 14, and 28 days and 3, 6, and 12 months after birth. Polymerase chain reaction-denaturing gradient gel electrophoresis (PCR-DGGE) was used to analyze the composition of intestinal microbiota, perform sequencing of dominant bacteria, and to analyze the changes in the composition of intestinal microbiota during infancy. RESULTS DGGE fingerprint showed that the composition of intestinal microbiota during infancy changed significantly over time after birth. The cloning and sequencing results indicated that Proteobacteria colonized the earliest, mainly the obligate aerobes Enterobacter and Pseudomonas, followed by the obligate anaerobes (Clostridium hathewayi and Veillonella parvula) and the facultative anaerobe Clostridium ramosum in Firmicutes, and Verrucomicrobia. Actinobacteria colonized the latest, mainly Bifidobacterium, and gradually became dominant bacteria. CONCLUSIONS During infancy, obligate aerobes colonize the intestinal tract the earliest, followed by obligate anaerobes and facultative anaerobes. Proteobacteria colonizes the earliest, followed by Firmicutes and Verrucomicrobia, and Actinobacteria, mainly Bifidobacterium, colonizes the latest.
Collapse
Affiliation(s)
- Shan-Shan Li
- Department of Nutrition, Food Safety and Toxicology, West China School of Public Health, Sichuan University, Chengdu 610041, China.
| | | | | | | | | | | | | |
Collapse
|
131
|
Timby N, Domellöf M, Holgerson PL, West CE, Lönnerdal B, Hernell O, Johansson I. Oral Microbiota in Infants Fed a Formula Supplemented with Bovine Milk Fat Globule Membranes - A Randomized Controlled Trial. PLoS One 2017; 12:e0169831. [PMID: 28099499 PMCID: PMC5242539 DOI: 10.1371/journal.pone.0169831] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 12/21/2016] [Indexed: 01/10/2023] Open
Abstract
Background In a recent study, supplementation of infant formula with milk fat globule membranes (MFGM) decreased the incidence of otitis media in infants <6 months of age. Objectives The aim of the present study was to characterize the oral microbiota in infants fed MFGM-supplemented formula and compare it to that of infants fed standard formula or breast milk. Methods In a prospective double-blinded randomized controlled trial, exclusively formula-fed infants <2 months of age were randomized to be fed experimental formula (EF, n = 80) with reduced energy and protein and supplemented with a bovine MFGM concentrate, or standard formula (SF, n = 80) until 6 months of age. A breast-fed reference (BFR, n = 80) group was also recruited. The oral microbiota was analyzed at 4 (n = 124) and 12 (n = 166) months of age using Illumina MiSeq multiplex sequencing and taxonomic resolution against the HOMD 16S rDNA database of oral bacteria. Results Species richness in the oral samples did not differ between the EF and SF groups, but partial least square modeling identified a few taxa that were significantly associated with being in either group, e.g. lower level of Moraxella catarrhalis in the EF group. Infants in the BFR group had significantly lower species richness at 4 months of age and their microbiota pattern differed markedly from the formula-fed groups. Conclusions Supplementation of infant formula with MFGM yielded moderate effects on the oral microbiome. Moraxella catarrhalis was less prevalent in infants fed EF than in those fed SF and may be associated with the decrease in otitis media seen in the same group.
Collapse
Affiliation(s)
- Niklas Timby
- Department of Clinical Sciences/Unit of Pediatrics, Umeå University, Umeå, Sweden
- * E-mail:
| | - Magnus Domellöf
- Department of Clinical Sciences/Unit of Pediatrics, Umeå University, Umeå, Sweden
| | | | - Christina E. West
- Department of Clinical Sciences/Unit of Pediatrics, Umeå University, Umeå, Sweden
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, United States of America
| | - Olle Hernell
- Department of Clinical Sciences/Unit of Pediatrics, Umeå University, Umeå, Sweden
| | - Ingegerd Johansson
- Department of Odontology/Unit of Cariology, Umeå University, Umeå, Sweden
| |
Collapse
|
132
|
Hill CJ, Lynch DB, Murphy K, Ulaszewska M, Jeffery IB, O'Shea CA, Watkins C, Dempsey E, Mattivi F, Tuohy K, Ross RP, Ryan CA, O' Toole PW, Stanton C. Evolution of gut microbiota composition from birth to 24 weeks in the INFANTMET Cohort. MICROBIOME 2017; 5:4. [PMID: 28095889 PMCID: PMC5240274 DOI: 10.1186/s40168-016-0213-y] [Citation(s) in RCA: 337] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/24/2016] [Indexed: 05/22/2023]
Abstract
BACKGROUND The gut is the most extensively studied niche of the human microbiome. The aim of this study was to characterise the initial gut microbiota development of a cohort of breastfed infants (n = 192) from 1 to 24 weeks of age. METHODS V4-V5 region 16S rRNA amplicon Illumina sequencing and, in parallel, bacteriological culture. The metabolomic profile of infant urine at 4 weeks of age was also examined by LC-MS. RESULTS Full-term (FT), spontaneous vaginally delivered (SVD) infants' microbiota remained stable at both phylum and genus levels during the 24-week period examined. FT Caesarean section (CS) infants displayed an increased faecal abundance of Firmicutes (p < 0.01) and lower abundance of Actinobacteria (p < 0.001) after the first week of life compared to FT-SVD infants. FT-CS infants gradually progressed to harbouring a microbiota closely resembling FT-SVD (which remained stable) by week 8 of life, which was maintained at week 24. The gut microbiota of preterm (PT) infants displayed a significantly greater abundance of Proteobacteria compared to FT infants (p < 0.001) at week 1. Metabolomic analysis of urine at week 4 indicated PT-CS infants have a functionally different metabolite profile than FT (both CS and SVD) infants. Co-inertia analysis showed co-variation between the urine metabolome and the faecal microbiota of the infants. Tryptophan and tyrosine metabolic pathways, as well as fatty acid and bile acid metabolism, were found to be affected by delivery mode and gestational age. CONCLUSIONS These findings confirm that mode of delivery and gestational age both have significant effects on early neonatal microbiota composition. There is also a significant difference between the metabolite profile of FT and PT infants. Prolonged breastfeeding was shown to have a significant effect on the microbiota composition of FT-CS infants at 24 weeks of age, but interestingly not on that of FT-SVD infants. Twins had more similar microbiota to one another than between two random infants, reflecting the influence of similarities in both host genetics and the environment on the microbiota..
Collapse
Affiliation(s)
- Cian J Hill
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Denise B Lynch
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Kiera Murphy
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Teagasc Moorepark Food Research Centre, Fermoy, Co. Cork, Ireland
| | - Marynka Ulaszewska
- Food Quality and Nutrition Department, Research and Innovation Centre, Fondazione Edmund Mach, San Michele All'adige, Italy
| | - Ian B Jeffery
- School of Microbiology, University College Cork, Cork, Ireland
| | - Carol Anne O'Shea
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - Claire Watkins
- Teagasc Moorepark Food Research Centre, Fermoy, Co. Cork, Ireland
| | - Eugene Dempsey
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - Fulvio Mattivi
- Food Quality and Nutrition Department, Research and Innovation Centre, Fondazione Edmund Mach, San Michele All'adige, Italy
| | - Kieran Tuohy
- Food Quality and Nutrition Department, Research and Innovation Centre, Fondazione Edmund Mach, San Michele All'adige, Italy
| | - R Paul Ross
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - C Anthony Ryan
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - Paul W O' Toole
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Institute, University College Cork, Cork, Ireland.
- Teagasc Moorepark Food Research Centre, Fermoy, Co. Cork, Ireland.
| |
Collapse
|
133
|
Davis EC, Wang M, Donovan SM. The role of early life nutrition in the establishment of gastrointestinal microbial composition and function. Gut Microbes 2017; 8:143-171. [PMID: 28068209 PMCID: PMC5390825 DOI: 10.1080/19490976.2016.1278104] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The development of the human infant intestinal microbiota is a sequential process that begins in utero and continues during the first 2 to 3 years of life. Microbial composition and diversity are shaped by host genetics and multiple environmental factors, of which diet is a principal contributor. An understanding of this process is of clinical importance as the microbiota acquired in early life influence gastrointestinal, immune and neural development, and reduced microbial diversity or dysbiosis during infancy is associated with disorders in infancy and later childhood. The goal of this article was to review the published literature that used culture-independent methods to describe the development of the gastrointestinal microbiota in breast- and formula-fed human infants as well as the impact of prebiotic and probiotic addition to infant formula, and the addition of solid foods.
Collapse
Affiliation(s)
- Erin C. Davis
- Division of Nutritional Sciences, Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL USA
| | - Mei Wang
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL USA
| | - Sharon M. Donovan
- Division of Nutritional Sciences, Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL USA,Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL USA,CONTACT Sharon M. Donovan University of Illinois, Department of Food Science and Human Nutrition, 339 Bevier Hall, 905 S. Goodwin Avenue, Urbana, IL 61801
| |
Collapse
|
134
|
Yasmin F, Tun HM, Konya TB, Guttman DS, Chari RS, Field CJ, Becker AB, Mandhane PJ, Turvey SE, Subbarao P, Sears MR, Scott JA, Dinu I, Kozyrskyj AL. Cesarean Section, Formula Feeding, and Infant Antibiotic Exposure: Separate and Combined Impacts on Gut Microbial Changes in Later Infancy. Front Pediatr 2017; 5:200. [PMID: 29018787 PMCID: PMC5622971 DOI: 10.3389/fped.2017.00200] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 08/31/2017] [Indexed: 12/11/2022] Open
Abstract
Established during infancy, our complex gut microbial community is shaped by medical interventions and societal preferences, such as cesarean section, formula feeding, and antibiotic use. We undertook this study to apply the significance analysis of microarrays (SAM) method to quantify changes in gut microbial composition during later infancy following the most common birth and postnatal exposures affecting infant gut microbial composition. Gut microbiota of 166 full-term infants in the Canadian Healthy Infant Longitudinal Development birth cohort were profiled using 16S high-throughput gene sequencing. Infants were placed into groups according to mutually exclusive combinations of birth mode (vaginal/cesarean birth), breastfeeding status (yes/no), and antibiotic use (yes/no) by 3 months of age. Based on repeated permutations of data and adjustment for the false discovery rate, the SAM statistic identified statistically significant changes in gut microbial abundance between 3 months and 1 year of age within each infant group. We observed well-known patterns of microbial phyla succession in later infancy (declining Proteobacteria; increasing Firmicutes and Bacteroidetes) following vaginal birth, breastfeeding, and no antibiotic exposure. Genus Lactobacillus, Roseburia, and Faecalibacterium species appeared in the top 10 increases to microbial abundance in these infants. Deviations from this pattern were evident among infants with other perinatal co-exposures; notably, the largest number of microbial species with unchanged abundance was seen in gut microbiota following early cessation of breastfeeding in infants. With and without antibiotic exposure, the absence of a breast milk diet by 3 months of age following vaginal birth yielded a higher proportion of unchanged abundance of Bacteroidaceae and Enterobacteriaceae in later infancy, and a higher ratio of unchanged Enterobacteriaceae to Alcaligenaceae microbiota. Gut microbiota of infants born vaginally and exclusively formula fed became less enriched with family Veillonellaceae and Clostridiaceae, showed unchanging levels of Ruminococcaceae, and exhibited a greater decline in the Rikenellaceae/Bacteroidaceae ratio compared to their breastfed, vaginally delivered counterparts. These changes were also evident in cesarean-delivered infants to a lesser extent. The clinical relevance of these trajectories of microbial change is that they culminate in taxon-specific abundances in the gut microbiota of later infancy, which we and others have observed to be associated with food sensitization.
Collapse
Affiliation(s)
- Farzana Yasmin
- Department of Public Health Sciences, University of Alberta, Edmonton, AB, Canada
| | - Hein Min Tun
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | | | - David S Guttman
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, ON, Canada
| | - Radha S Chari
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB, Canada
| | - Allan B Becker
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Piush J Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Stuart E Turvey
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Padmaja Subbarao
- Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Malcolm R Sears
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | | | - James A Scott
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Irina Dinu
- Department of Public Health Sciences, University of Alberta, Edmonton, AB, Canada
| | - Anita L Kozyrskyj
- Department of Public Health Sciences, University of Alberta, Edmonton, AB, Canada.,Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.,Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
135
|
Glycosulfatase-Encoding Gene Cluster in Bifidobacterium breve UCC2003. Appl Environ Microbiol 2016; 82:6611-6623. [PMID: 27590817 DOI: 10.1128/aem.02022-16] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 08/26/2016] [Indexed: 12/11/2022] Open
Abstract
Bifidobacteria constitute a specific group of commensal bacteria typically found in the gastrointestinal tract (GIT) of humans and other mammals. Bifidobacterium breve strains are numerically prevalent among the gut microbiota of many healthy breastfed infants. In the present study, we investigated glycosulfatase activity in a bacterial isolate from a nursling stool sample, B. breve UCC2003. Two putative sulfatases were identified on the genome of B. breve UCC2003. The sulfated monosaccharide N-acetylglucosamine-6-sulfate (GlcNAc-6-S) was shown to support the growth of B. breve UCC2003, while N-acetylglucosamine-3-sulfate, N-acetylgalactosamine-3-sulfate, and N-acetylgalactosamine-6-sulfate did not support appreciable growth. By using a combination of transcriptomic and functional genomic approaches, a gene cluster designated ats2 was shown to be specifically required for GlcNAc-6-S metabolism. Transcription of the ats2 cluster is regulated by a repressor open reading frame kinase (ROK) family transcriptional repressor. This study represents the first description of glycosulfatase activity within the Bifidobacterium genus. IMPORTANCE Bifidobacteria are saccharolytic organisms naturally found in the digestive tract of mammals and insects. Bifidobacterium breve strains utilize a variety of plant- and host-derived carbohydrates that allow them to be present as prominent members of the infant gut microbiota as well as being present in the gastrointestinal tract of adults. In this study, we introduce a previously unexplored area of carbohydrate metabolism in bifidobacteria, namely, the metabolism of sulfated carbohydrates. B. breve UCC2003 was shown to metabolize N-acetylglucosamine-6-sulfate (GlcNAc-6-S) through one of two sulfatase-encoding gene clusters identified on its genome. GlcNAc-6-S can be found in terminal or branched positions of mucin oligosaccharides, the glycoprotein component of the mucous layer that covers the digestive tract. The results of this study provide further evidence of the ability of this species to utilize mucin-derived sugars, a trait which may provide a competitive advantage in both the infant gut and adult gut.
Collapse
|
136
|
Tannock GW, Lee PS, Wong KH, Lawley B. Why Don't All Infants Have Bifidobacteria in Their Stool? Front Microbiol 2016; 7:834. [PMID: 27303402 PMCID: PMC4886621 DOI: 10.3389/fmicb.2016.00834] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/17/2016] [Indexed: 01/03/2023] Open
Affiliation(s)
- Gerald W Tannock
- Department of Microbiology and Immunology, University of OtagoDunedin, New Zealand; Riddet Centre for Research Excellence, Massey UniversityPalmerston North, New Zealand
| | - Pheng Soon Lee
- Mead Johnson NutritionSingapore, Singapore; Department of Human Nutrition, University of OtagoDunedin, New Zealand
| | | | - Blair Lawley
- Department of Microbiology and Immunology, University of Otago Dunedin, New Zealand
| |
Collapse
|
137
|
Bacterial Succession in the Broiler Gastrointestinal Tract. Appl Environ Microbiol 2016; 82:2399-2410. [PMID: 26873323 DOI: 10.1128/aem.02549-15] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 02/06/2016] [Indexed: 01/13/2023] Open
Abstract
A feeding trial was performed with broilers receiving a diet of wheat-based feed (WBF), maize-based feed (MBF), or maize-based concentrates supplemented with 15% or 30% crimped kernel maize silage (CKMS-15 or CKMS-30, respectively). The aim of the study was to investigate the bacterial community compositions of the crop, gizzard, ileum, and cecum contents in relation to the feeding strategy and age (8, 15, 22, 25, 29, or 36 days). Among the four dietary treatments, bacterial diversity was analyzed for MBF and CKMS-30 by 454 pyrosequencing of the 16S rRNA gene. Since the diets had no significant influence on bacterial diversity, data were pooled for downstream analysis. With increasing age, a clear succession of bacterial communities and increased bacterial diversity were observed.Lactobacillaceae(belonging mainly to the genus Lactobacillus) represented most of the Firmicutesat all ages and in all segments of the gut except the cecum. The development of a "mature" microbiota in broilers occurred during the period from days 15 to 22. Striking increases in the relative abundances of Lactobacillus salivarius(17 to 36%) and clostridia (11 to 18%), and a concomitant decrease in the relative abundance of Lactobacillus reuteri, were found in the ileum after day 15. The concentration of deconjugated bile salts increased in association with the increased populations of L. salivarius and clostridia. Both L. salivarius and clostridia deconjugate bile acids, and increases in the abundances of these bacteria might be associated with growth reduction and gastrointestinal (GI) disorders occurring in the critical period of broiler life between days 20 and 30.
Collapse
|
138
|
Katayama T. Host-derived glycans serve as selected nutrients for the gut microbe: human milk oligosaccharides and bifidobacteria†. Biosci Biotechnol Biochem 2016; 80:621-32. [DOI: 10.1080/09168451.2015.1132153] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Abstract
Lactation is a common feeding strategy of eutherian mammals, but its functions go beyond feeding the neonates. Ever since Tissier isolated bifidobacteria from the stool of breast-fed infants, human milk has been postulated to contain compounds that selectively stimulate the growth of bifidobacteria in intestines. However, until relatively recently, there have been no reports to link human milk compound(s) with bifidobacterial physiology. Over the past decade, successive studies have demonstrated that infant-gut-associated bifidobacteria are equipped with genetic and enzymatic toolsets dedicated to assimilation of host-derived glycans, especially human milk oligosaccharides (HMOs). Among gut microbes, the presence of enzymes required for degrading HMOs with type-1 chains is essentially limited to infant-gut-associated bifidobacteria, suggesting HMOs serve as selected nutrients for the bacteria. In this study, I shortly discuss the research on bifidobacteria and HMOs from a historical perspective and summarize the roles of bifidobacterial enzymes in the assimilation of HMOs with type-1 chains. Based on this overview, I suggest the co-evolution between bifidobacteria and human beings mediated by HMOs.
Collapse
Affiliation(s)
- Takane Katayama
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Faculty of Bioresources and Environmental Sciences, Ishikawa Prefectural University, Nonoichi, Japan
| |
Collapse
|
139
|
Madan JC, Hoen AG, Lundgren SN, Farzan SF, Cottingham KL, Morrison HG, Sogin ML, Li H, Moore JH, Karagas MR. Association of Cesarean Delivery and Formula Supplementation With the Intestinal Microbiome of 6-Week-Old Infants. JAMA Pediatr 2016; 170:212-9. [PMID: 26752321 PMCID: PMC4783194 DOI: 10.1001/jamapediatrics.2015.3732] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
IMPORTANCE The intestinal microbiome plays a critical role in infant development, and delivery mode and feeding method (breast milk vs formula) are determinants of its composition. However, the importance of delivery mode beyond the first days of life is unknown, and studies of associations between infant feeding and microbiome composition have been generally limited to comparisons between exclusively breastfed and formula-fed infants, with little consideration given to combination feeding of both breast milk and formula. OBJECTIVE To examine the associations of delivery mode and feeding method with infant intestinal microbiome composition at approximately 6 weeks of life. DESIGN, SETTING, AND PARTICIPANTS Prospective observational study of 102 infants followed up as part of a US pregnancy cohort study. EXPOSURES Delivery mode was abstracted from delivery medical records, and feeding method prior to the time of stool collection was ascertained through detailed questionnaires. MAIN OUTCOMES AND MEASURES Stool microbiome composition was characterized using next-generation sequencing of the 16S rRNA gene. RESULTS There were 102 infants (mean gestational age, 39.7 weeks; range, 37.1-41.9 weeks) included in this study, of whom 70 were delivered vaginally and 32 by cesarean delivery. In the first 6 weeks of life, 70 were exclusively breastfed, 26 received combination feeding, and 6 were exclusively formula fed. We identified independent associations between microbial community composition and both delivery mode (P< .001; Q < .001) and feeding method (P = .01; Q < .001). Differences in microbial community composition between vaginally delivered infants and infants delivered by cesarean birth were equivalent to or significantly larger than those between feeding groups (P = .003). Bacterial communities associated with combination feeding were more similar to those associated with exclusive formula feeding than exclusive breastfeeding (P = .002). We identified 6 individual bacterial genera that were differentially abundant between delivery mode and feeding groups. CONCLUSIONS AND RELEVANCE The infant intestinal microbiome at approximately 6 weeks of age is significantly associated with both delivery mode and feeding method, and the supplementation of breast milk feeding with formula is associated with a microbiome composition that resembles that of infants who are exclusively formula fed. These results may inform feeding choices and shed light on the mechanisms behind the lifelong health consequences of delivery and infant feeding modalities.
Collapse
Affiliation(s)
- Juliette C. Madan
- Division of Neonatology, Department of Pediatrics, Children’s Hospital at Dartmouth, Lebanon, NH, USA,Children’s Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, New Hampshire, USA
| | - Anne G. Hoen
- Children’s Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, New Hampshire, USA,Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, USA,Computational Genetics Laboratory, Geisel School of Medicine at Dartmouth, Hanover, NH, USA,Corresponding author: Anne G. Hoen, PhD, Department of Epidemiology, Geisel School of Medicine at Dartmouth. 1 Medical Center Drive, Lebanon, NH 03756. 603-653-6087.
| | - Sara N. Lundgren
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Shohreh F. Farzan
- Children’s Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, New Hampshire, USA,Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Kathryn L. Cottingham
- Children’s Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, New Hampshire, USA,Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Hilary G. Morrison
- Josephine Bay Paul Center, Marine Biological Laboratory, Woods Hole, MA, USA
| | - Mitchell L. Sogin
- Josephine Bay Paul Center, Marine Biological Laboratory, Woods Hole, MA, USA
| | - Hongzhe Li
- Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason H. Moore
- Computational Genetics Laboratory, Geisel School of Medicine at Dartmouth, Hanover, NH, USA,Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Margaret R. Karagas
- Children’s Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, New Hampshire, USA,Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, USA,Center for Molecular Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| |
Collapse
|
140
|
Simeoni U, Berger B, Junick J, Blaut M, Pecquet S, Rezzonico E, Grathwohl D, Sprenger N, Brüssow H, Szajewska H, Bartoli J, Brevaut‐Malaty V, Borszewska‐Kornacka M, Feleszko W, François P, Gire C, Leclaire M, Maurin J, Schmidt S, Skórka A, Squizzaro C, Verdot J. Gut microbiota analysis reveals a marked shift to bifidobacteria by a starter infant formula containing a synbiotic of bovine milk‐derived oligosaccharides and
B
ifidobacterium animalis
subsp.
lactis
CNCM I
‐3446. Environ Microbiol 2016; 18:2185-95. [DOI: 10.1111/1462-2920.13144] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 11/03/2015] [Accepted: 11/19/2015] [Indexed: 01/15/2023]
Affiliation(s)
- Umberto Simeoni
- Pédiatrie (PED) Centre Hospitalier Universitaire Vaudois Lausanne Switzerland
| | | | - Jana Junick
- Gastrointestinal Microbiology German Institute of Human Nutrition Postdam‐Rehbrücke Nuthetal Germany
| | - Michael Blaut
- Gastrointestinal Microbiology German Institute of Human Nutrition Postdam‐Rehbrücke Nuthetal Germany
| | - Sophie Pecquet
- Nestlé Nutrition Clinical Development Unit Nestec Ltd Vevey Switzerland
| | | | | | | | | | - Hania Szajewska
- Department of Paediatrics Medical University of Warsaw Warsaw Poland
| | | | | | | | - W. Feleszko
- Department of Paediatrics Medical University of Warsaw Warsaw Poland
| | | | - C. Gire
- Hôpital Nord Marseille France
| | | | | | | | - A. Skórka
- Department of Paediatrics Medical University of Warsaw Warsaw Poland
| | | | | | | |
Collapse
|
141
|
Sagheddu V, Patrone V, Miragoli F, Puglisi E, Morelli L. Infant Early Gut Colonization by Lachnospiraceae: High Frequency of Ruminococcus gnavus. Front Pediatr 2016; 4:57. [PMID: 27313996 PMCID: PMC4889575 DOI: 10.3389/fped.2016.00057] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 05/19/2016] [Indexed: 12/28/2022] Open
Abstract
Lachnospiraceae is a bacterial family usually isolated from human and mammalian intestinal microbiota. However, its presence and role in the infant microbiota is not fully elucidated. This may be due to the strictly anaerobic behavior of its members that hampers the possibility of culture-dependent enumeration. Here, we report on the presence of this bacterial group, using biomolecular techniques, in stool samples from 25 babies aged between 1 and 24 months. Denaturing gradient gel electrophoresis (DGGE) was used as a first detection step, and data were confirmed by quantitative PCR (qPCR). The DGGE showed the presence of Lachnospiraceae in infant fecal specimens and indicated the prevalence of Ruminococcus gnavus (R. gnavus). The qPCR confirmed the presence of the Clostridium XVIa group, Blautia genus, and R. gnavus, which are the main members of this family. We detected R. gnavus in 22 of 25 (88%) samples with a qPCR probe assay. Despite the difficulties associated with their detection and enumeration, Lachnospiraceae, and in particular R. gnavus, should be included in future studies on the infant microbiota composition.
Collapse
Affiliation(s)
- Valeria Sagheddu
- Facoltà di Scienze Agrarie, Alimentari e Ambientali, Istituto di Microbiologia, Università Cattolica del Sacro Cuore , Piacenza , Italy
| | - Vania Patrone
- Facoltà di Scienze Agrarie, Alimentari e Ambientali, Istituto di Microbiologia, Università Cattolica del Sacro Cuore , Piacenza , Italy
| | - Francesco Miragoli
- Facoltà di Scienze Agrarie, Alimentari e Ambientali, Istituto di Microbiologia, Università Cattolica del Sacro Cuore , Piacenza , Italy
| | - Edoardo Puglisi
- Facoltà di Scienze Agrarie, Alimentari e Ambientali, Istituto di Microbiologia, Università Cattolica del Sacro Cuore , Piacenza , Italy
| | - Lorenzo Morelli
- Facoltà di Scienze Agrarie, Alimentari e Ambientali, Istituto di Microbiologia, Università Cattolica del Sacro Cuore , Piacenza , Italy
| |
Collapse
|
142
|
Groer MW, Gregory KE, Louis-Jacques A, Thibeau S, Walker WA. The very low birth weight infant microbiome and childhood health. ACTA ACUST UNITED AC 2015; 105:252-64. [PMID: 26663857 DOI: 10.1002/bdrc.21115] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This review describes current understandings about the nature of the very low birth weight infant (VLBW) gut microbiome. VLBW infants often experience disruptive pregnancies and births, and prenatal factors can influence the maturity of the gut and immune system, and disturb microbial balance and succession. Many VLBWs experience rapid vaginal or Caesarean births. After birth these infants often have delays in enteral feeding, and many receive little or no mother's own milk. Furthermore the stressors of neonatal life in the hospital environment, common use of antibiotics, invasive procedures and maternal separation can contribute to dysbiosis. These infants experience gastrointestinal dysfunction, sepsis, transfusions, necrotizing enterocolitis, oxygen toxicity, and other pathophysiological conditions that affect the normal microbiota. The skin is susceptible to dysbiosis, due to its fragility and contact with NICU organisms. Dysbiosis in early life may resolve but little is known about the timing of the development of the signature gut microbiome in VLBWs. Dysbiosis has been associated with a number of physical and behavioral problems, including autism spectrum disorders, allergy and asthma, gastrointestinal disease, obesity, depression, and anxiety. Dysbiosis may be prevented or ameliorated in part by prenatal care, breast milk feeding, skin to skin contact, use of antibiotics only when necessary, and vigilance during infancy and early childhood.
Collapse
Affiliation(s)
- Maureen W Groer
- Morsani College of Medicine, University of South Florida College of Nursing, Tampa, Florida
| | - Katherine E Gregory
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Department of Nursing, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Adetola Louis-Jacques
- Morsani College of Medicine, University of South Florida College of Nursing, Tampa, Florida
| | | | - W Allan Walker
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
143
|
Xu M, Wang Y, Dai Z, Zhang Y, Li Y, Wang J. Comparison of growth and nutritional status in infants receiving goat milk-based formula and cow milk-based formula: a randomized, double-blind study. Food Nutr Res 2015; 59:28613. [PMID: 26652603 PMCID: PMC4676839 DOI: 10.3402/fnr.v59.28613] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 11/12/2015] [Accepted: 11/12/2015] [Indexed: 11/28/2022] Open
Abstract
Objective To compare the growth and nutritional status of infants fed goat milk–based formula (GMF) and cow milk–based formula (CMF). Methods The study was conducted in Beijing, China. It was a double-blind randomized controlled trial. A total of 79 infants aged 0–3 months old were recruited and randomized in GMF or CMF group. The infants were fed the allocated formula to 6 months. The weight, length, and head circumference were measured at the enrolment, 3 and 6 months. The start time and types of solid food were recorded. Blood elements, urinal, and fecal parameters were also tested. Results The average weight of infants in the GMF group (mean±SD) was 4.67±0.99 kg and in the CMF group 4.73±1.10 kg at enrolment, and 8.75±0.98 kg (GMF) and 8.92±0.88 kg (CMF) at 6 months. There were no differences in the adjusted intention-to-treat analyses of weight, length, head circumference, and BMI z-scores between the two formula-fed groups over the 6-month study. Similarly, there were no remarkable differences in the timing and types of solid food, blood elements, urinal, and feces parameters, between the GMF and CMF group. No group differences have been shown in bowel motion consistency, duration of crying, ease of settling, or frequency of adverse events. Conclusions GMF-provided growth and nutritional outcomes did not differ from those provided by CMF.
Collapse
Affiliation(s)
- Meihong Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing, China
| | - Yibin Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing, China
| | - Zhiyong Dai
- Ausnutria Hyproca Dairy Group BV, Changsha, China
| | | | - Yong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing, China
| | - Junbo Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing, China;
| |
Collapse
|
144
|
Affiliation(s)
- Anita L Kozyrskyj
- a Department of Pediatrics , University of Alberta , Edmonton , Canada
| |
Collapse
|
145
|
Bifidobacterium breve alters immune function and ameliorates DSS-induced inflammation in weanling rats. Pediatr Res 2015; 78:407-16. [PMID: 26083761 DOI: 10.1038/pr.2015.115] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/13/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND Bifidobacterium breve M-16V (M16V) is a probiotic bacterial strain with a long tradition of use in neonatal intensive care units in some countries. Previous study showed that the effects of M16V administration on gene expression were greater during the weaning period than in the neonatal period and were greater in the colon than in the small intestine and spleen, suggesting that M16V has anti-inflammatory effects. In this study, we evaluated the effects of inflammation during the weaning period and the effects of M16V on normal and inflammatory conditions. METHODS From postnatal day (PD) 21 to 34, weanling rats were administered of 2.5 × 10(9) of M16V daily, and colitis was induced by administration of 2% dextran sulfate sodium from PD28 to 35. Colitis severity, immune function, and microbiota were investigated. RESULTS Colitis caused a reduction in body weight gain, colon shortening, poor nutritional status, anemia, changes in blood and spleen lymphocyte populations, spleen T-cell malfunctions, and alterations in colon microbiota. M16V administration improved some but not all of the changes induced by colitis. CONCLUSION M16V could suppress inflammation and, therefore, can be considered a safe strain to use not only during the neonatal period but also the weaning period.
Collapse
|
146
|
Tailford LE, Crost EH, Kavanaugh D, Juge N. Mucin glycan foraging in the human gut microbiome. Front Genet 2015; 6:81. [PMID: 25852737 PMCID: PMC4365749 DOI: 10.3389/fgene.2015.00081] [Citation(s) in RCA: 565] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 02/16/2015] [Indexed: 12/21/2022] Open
Abstract
The availability of host and dietary carbohydrates in the gastrointestinal (GI) tract plays a key role in shaping the structure-function of the microbiota. In particular, some gut bacteria have the ability to forage on glycans provided by the mucus layer covering the GI tract. The O-glycan structures present in mucin are diverse and complex, consisting predominantly of core 1-4 mucin-type O-glycans containing α- and β- linked N-acetyl-galactosamine, galactose and N-acetyl-glucosamine. These core structures are further elongated and frequently modified by fucose and sialic acid sugar residues via α1,2/3/4 and α2,3/6 linkages, respectively. The ability to metabolize these mucin O-linked oligosaccharides is likely to be a key factor in determining which bacterial species colonize the mucosal surface. Due to their proximity to the immune system, mucin-degrading bacteria are in a prime location to influence the host response. However, despite the growing number of bacterial genome sequences available from mucin degraders, our knowledge on the structural requirements for mucin degradation by gut bacteria remains fragmented. This is largely due to the limited number of functionally characterized enzymes and the lack of studies correlating the specificity of these enzymes with the ability of the strain to degrade and utilize mucin and mucin glycans. This review focuses on recent findings unraveling the molecular strategies used by mucin-degrading bacteria to utilize host glycans, adapt to the mucosal environment, and influence human health.
Collapse
Affiliation(s)
| | | | | | - Nathalie Juge
- The Gut Health and Food Safety Institute Strategic Programme, Institute of Food ResearchNorwich, UK
| |
Collapse
|
147
|
Azad MB, Konya T, Guttman DS, Field CJ, Sears MR, HayGlass KT, Mandhane PJ, Turvey SE, Subbarao P, Becker AB, Scott JA, Kozyrskyj AL. Infant gut microbiota and food sensitization: associations in the first year of life. Clin Exp Allergy 2015; 45:632-43. [DOI: 10.1111/cea.12487] [Citation(s) in RCA: 276] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/22/2014] [Accepted: 12/21/2014] [Indexed: 12/20/2022]
Affiliation(s)
- M. B. Azad
- Department of Pediatrics; School of Public Health; University of Alberta; Edmonton AB Canada
- Department of Pediatrics & Child Health; Children's Hospital Research Institute of Manitoba; University of Manitoba; Winnipeg MB Canada
| | - T. Konya
- Dalla Lana School of Public Health; University of Toronto; Toronto ON Canada
| | - D. S. Guttman
- Centre for the Analysis of Genome Evolution and Function; University of Toronto; Toronto ON Canada
| | - C. J. Field
- Department of Agricultural, Food & Nutritional Science; University of Alberta; Edmonton AB Canada
| | - M. R. Sears
- Department of Medicine; McMaster University; Hamilton ON Canada
| | - K. T. HayGlass
- Department of Immunology; University of Manitoba; Winnipeg MB Canada
| | - P. J. Mandhane
- Department of Pediatrics; School of Public Health; University of Alberta; Edmonton AB Canada
| | - S. E. Turvey
- Department of Pediatrics, Child & Family Research Institute; BC Children's Hospital; University of British Columbia; Vancouver BC Canada
| | - P. Subbarao
- Department of Pediatrics; Hospital for Sick Children; University of Toronto; Toronto ON Canada
| | - A. B. Becker
- Department of Pediatrics & Child Health; Children's Hospital Research Institute of Manitoba; University of Manitoba; Winnipeg MB Canada
| | - J. A. Scott
- Dalla Lana School of Public Health; University of Toronto; Toronto ON Canada
| | - A. L. Kozyrskyj
- Department of Pediatrics; School of Public Health; University of Alberta; Edmonton AB Canada
- Department of Community Health Sciences; University of Manitoba; Winnipeg MB Canada
| | | |
Collapse
|
148
|
Thompson AL, Monteagudo-Mera A, Cadenas MB, Lampl ML, Azcarate-Peril MA. Milk- and solid-feeding practices and daycare attendance are associated with differences in bacterial diversity, predominant communities, and metabolic and immune function of the infant gut microbiome. Front Cell Infect Microbiol 2015; 5:3. [PMID: 25705611 PMCID: PMC4318912 DOI: 10.3389/fcimb.2015.00003] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/05/2015] [Indexed: 01/14/2023] Open
Abstract
The development of the infant intestinal microbiome in response to dietary and other exposures may shape long-term metabolic and immune function. We examined differences in the community structure and function of the intestinal microbiome between four feeding groups, exclusively breastfed infants before introduction of solid foods (EBF), non-exclusively breastfed infants before introduction of solid foods (non-EBF), EBF infants after introduction of solid foods (EBF+S), and non-EBF infants after introduction of solid foods (non-EBF+S), and tested whether out-of-home daycare attendance was associated with differences in relative abundance of gut bacteria. Bacterial 16S rRNA amplicon sequencing was performed on 49 stool samples collected longitudinally from a cohort of 9 infants (5 male, 4 female). PICRUSt metabolic inference analysis was used to identify metabolic impacts of feeding practices on the infant gut microbiome. Sequencing data identified significant differences across groups defined by feeding and daycare attendance. Non-EBF and daycare-attending infants had higher diversity and species richness than EBF and non-daycare attending infants. The gut microbiome of EBF infants showed increased proportions of Bifidobacterium and lower abundance of Bacteroidetes and Clostridiales than non-EBF infants. PICRUSt analysis indicated that introduction of solid foods had a marginal impact on the microbiome of EBF infants (24 enzymes overrepresented in EBF+S infants). In contrast, over 200 bacterial gene categories were overrepresented in non-EBF+S compared to non-EBF infants including several bacterial methyl-accepting chemotaxis proteins (MCP) involved in signal transduction. The identified differences between EBF and non-EBF infants suggest that breast milk may provide the gut microbiome with a greater plasticity (despite having a lower phylogenetic diversity) that eases the transition into solid foods.
Collapse
Affiliation(s)
- Amanda L. Thompson
- Department of Anthropology, University of North CarolinaChapel Hill, NC, USA
| | - Andrea Monteagudo-Mera
- Microbiome Core Facility, Center for Gastrointestinal Biology and Disease, University of North CarolinaChapel Hill, NC, USA
| | - Maria B. Cadenas
- Microbiome Core Facility, Center for Gastrointestinal Biology and Disease, University of North CarolinaChapel Hill, NC, USA
| | - Michelle L. Lampl
- Department of Anthropology and Center for the Study of Human Health, Emory UniversityAtlanta, GA, USA
| | - M. A. Azcarate-Peril
- Microbiome Core Facility, Center for Gastrointestinal Biology and Disease, University of North CarolinaChapel Hill, NC, USA
- Department of Cell Biology and Physiology, School of Medicine, University of North CarolinaChapel Hill, NC, USA
| |
Collapse
|
149
|
Lee SA, Lim JY, Kim BS, Cho SJ, Kim NY, Kim OB, Kim Y. Comparison of the gut microbiota profile in breast-fed and formula-fed Korean infants using pyrosequencing. Nutr Res Pract 2014; 9:242-8. [PMID: 26060535 PMCID: PMC4460055 DOI: 10.4162/nrp.2015.9.3.242] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 08/20/2014] [Accepted: 08/21/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND/OBJECTIVES Feeding in infancy is the most significant determinant of the intestinal microbiota in early life. The aim of this study was to determine the gut microbiota of Korean infants and compare the microbiota obtained between breast-fed and formula-fed Korean infants. SUBJECTS/METHODS We analyzed the microbial communities in fecal samples collected from twenty 4-week old Korean (ten samples in each breast-fed or formula-fed) infants using pyrosequencing. RESULTS The fecal microbiota of the 4-week-old Korean infants consisted of the three phyla Actinobacteria, Firmicutes, and Proteobacteria. In addition, five species, including Bifidocbacterium longum, Streptococcus salivarius, Strepotococcus lactarius, Streptococcus pseudopneumoniae, and Lactobacillus gasseri were common commensal intestinal microbiota in all infants. The predominant intestinal microbiota in the breast-fed infants (BFI) included the phylum Actinobacteria (average 70.55%), family Bifidobacteriacea (70.12%), genus Bifidobacterium (70.03%) and species Bifidobacterium longum (69.96%). In the microbiota from the formula-fed infants (FFI), the proportion of the phylum Actinobacteria (40.68%) was less, whereas the proportions of Firmicutes (45.38%) and Proteobacteria (13.85%) as well as the diversity of each taxonomic level were greater, compared to those of the BFI. The probiotic species found in the 4-week-old Korean infants were Bifidobacterium longum, Streptococcus salivarius, and Lactobacillus gasseri. These probiotic species accounted for 93.81% of the microbiota from the BFI, while only 63.80% of the microbiota from the FFI. In particular, B. longum was more abundant in BFI (69.96%) than in FFI (34.17%). CONCLUSIONS Breast milk supports the growth of B. longum and inhibits others. To the best of our knowledge, this study was the first attempt to analyze the gut microbiota of healthy Korean infants according to the feeding type using pyrosequencing. Our data can be used as a basis for further studies to investigate the development of intestinal microbiota with aging and disease status.
Collapse
Affiliation(s)
- Sang A Lee
- The Graduate School of Clinical Health Sciences, Ewha Womans University, Seoul 120-750, Korea
| | - Ji Ye Lim
- Department of Nutritional Science and Food Management, Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul 120-750, Korea
| | - Bong-Soo Kim
- ChunLab, Inc. Seoul 151-742, Korea. ; Department of Life Science, Hallym University, Gangwon 200-702, Korea
| | - Su Jin Cho
- Department of Pediatrics, School of Medicine, Ewha Womans University, Seoul 158-710, Korea
| | - Nak Yon Kim
- Arante Women's Hospital, Seoul 150-836, Korea
| | - Ok Bin Kim
- Department of Life Science, Global Top 5 Research Program, Ewha Womans University, Seoul 120-750, Korea
| | - Yuri Kim
- The Graduate School of Clinical Health Sciences, Ewha Womans University, Seoul 120-750, Korea. ; Department of Nutritional Science and Food Management, Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul 120-750, Korea
| |
Collapse
|
150
|
Hesla HM, Stenius F, Jäderlund L, Nelson R, Engstrand L, Alm J, Dicksved J. Impact of lifestyle on the gut microbiota of healthy infants and their mothers - the ALADDIN birth cohort. FEMS Microbiol Ecol 2014; 90:791-801. [DOI: 10.1111/1574-6941.12434] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 09/29/2014] [Accepted: 09/30/2014] [Indexed: 12/17/2022] Open
Affiliation(s)
- Helena M. Hesla
- Sachs' Children and Youth Hospital; Södersjukhuset; Stockholm Sweden
- Department of Clinical Science and Education Södersjukhuset; Karolinska Institutet; Stockholm Sweden
| | - Fredrik Stenius
- Sachs' Children and Youth Hospital; Södersjukhuset; Stockholm Sweden
- Department of Clinical Science and Education Södersjukhuset; Karolinska Institutet; Stockholm Sweden
| | - Lotta Jäderlund
- Department of Microbiology; Swedish University of Agricultural Sciences; Uppsala Sweden
| | - Ronald Nelson
- Department of Clinical Sciences; Swedish University of Agricultural Sciences; Uppsala Sweden
| | - Lars Engstrand
- Department of Microbiology, Tumour and Cell Biology; Karolinska Institutet; Stockholm Sweden
| | - Johan Alm
- Sachs' Children and Youth Hospital; Södersjukhuset; Stockholm Sweden
- Department of Clinical Science and Education Södersjukhuset; Karolinska Institutet; Stockholm Sweden
| | - Johan Dicksved
- Department of Microbiology; Swedish University of Agricultural Sciences; Uppsala Sweden
- Department of Animal Nutrition and Management; Swedish University of Agricultural Sciences; Uppsala Sweden
| |
Collapse
|