Dysregulated metabolism and behaviors by disrupting gut microbiota in prenatal and neonatal mice

2021 ◽  
Vol 92 (1) ◽  
Author(s):  
Guofeng Han ◽  
Takuma Nishigawa ◽  
Hiromi Ikeda ◽  
Mizuki Hamada ◽  
Hui Yang ◽  
...  
2019 ◽  
Vol 149 (2) ◽  
pp. 344-353 ◽  
Author(s):  
Hirohisa Izumi ◽  
Tatsuya Ehara ◽  
Hirosuke Sugahara ◽  
Takeshi Matsubara ◽  
Eri Mitsuyama ◽  
...  

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.


2019 ◽  
Vol 10 (2) ◽  
pp. 925-937 ◽  
Author(s):  
Ye Sun ◽  
Weitao Geng ◽  
Yajing Pan ◽  
Jinju Wang ◽  
Ping Xiao ◽  
...  

ZW3 regulated the biomarkers and behaviors associated with tryptophan metabolism, the HPA axis, and the immune system in depressed mice by regulating the gut microbiota.


2016 ◽  
Vol 116 (2) ◽  
pp. 270-278 ◽  
Author(s):  
Tatsuya Ehara ◽  
Hirohisa Izumi ◽  
Muneya Tsuda ◽  
Yuki Nakazato ◽  
Hiroshi Iwamoto ◽  
...  

AbstractIt is important to provide formula-fed infants with a bifidobacteria-enriched gut microbiota similar to those of breastfed infants to ensure intestinal health. Prebiotics, such as certain oligosaccharides, are a useful solution to this problem, but the combinational benefits of these oligosaccharides have not been evaluated. This study investigated the benefits of oligosaccharide combinations and screened for an optimal combination of oligosaccharides to promote healthy gut microbiota of formula-fed infants. In vitro and in vivo experiments were performed to assess the bifidogenic effects of lactulose (LAC) alone and LAC combined with raffinose (RAF) and/or galacto-oligosaccharide (GOS), using a mixed culture model and neonatal mice orally administered with these oligosaccharides and Bifidobacterium breve. In the in vitro culture model, the combination of the three oligosaccharides (LAC–RAF–GOS) significantly increased cell numbers of B. breve and Bifidobacterium longum (P<0·05) compared with either LAC alone or the combination of two oligosaccharides, and resulted in the production of SCFA under anaerobic conditions. In the in vivo experiment, the LAC–RAF–GOS combination significantly increased cell numbers of B. breve and Bacteroidetes in the large intestinal content (P<0·05) and increased acetate concentrations in the caecal content and serum of neonatal mice. Genes related to metabolism and immune responses were differentially expressed in the liver and large intestine of mice administered with LAC–RAF–GOS. These results indicate a synergistic effect of the LAC–RAF–GOS combination on the growth of bifidobacteria and reveal possible benefits of this combination to the gut microbiota and health of infants.


2019 ◽  
Author(s):  
Kevin R Hughes ◽  
Z Schofield ◽  
MJ Dalby ◽  
S Caim ◽  
L Chalklen ◽  
...  

AbstractThe gut microbiota plays a crucial role in regulating and maintaining the epithelial barrier, particularly during early life. Notably, patients with chronic intestinal inflammation have a dysregulated process of renewal and replenishment of the intestinal epithelial cell (IEC) barrier, which is linked to disturbances in the gut microbiota. To date, there are no studies focussed on understanding the impact of inflammatory cell shedding events during the early life developmental window, and which host and microbial factors mediate these responses. Here we sought to determine pathological cell shedding outcomes throughout the postnatal developmental period (day 14, 21, 29 and week 8). Surprisingly neonatal mice (day 14 and 21) were highly refractory to induction of cell shedding after intraperitoneal administration of LPS, with day 29 mice showing strong pathological responses, more similar to those observed in adult mice. These differential responses were not linked to defects in the cellular mechanisms and pathways known to regulate cell shedding responses, although we did observe that neonatal mice had elevated anti-inflammatory (IL-10) responses. Notably, when we profiled microbiota and metabolites from these mice, we observed significant alterations. Neonatal mice had high relative abundances of Streptococcus, Escherichia and Enterococcus and increased primary bile acids. In contrast, older mice were dominated by Candidatus Arthromitus, Alistipes and Lachnoclostridium, and had increased concentrations of SCFAs and methyamines. Faecal microbiota transplant (FMT) and antibiotic studies confirmed the importance of early life gut microbiota in cell shedding responses. In these studies, neonates treated with antibiotics restored LPS-induced small intestinal cell shedding, whereas adult FMT alone had no effect. Our findings further support the importance of the early life window for microbiota-epithelial interactions in the presence of inflammatory stimuli and highlight areas for further investigation to probe underlying mechanisms to drive therapeutic development within the context of chronic inflammatory intestinal diseases.


2019 ◽  
Vol 64 (2) ◽  
pp. 268-275 ◽  
Author(s):  
Mohamed Mammeri ◽  
Aurélie Chevillot ◽  
Myriam Thomas ◽  
Christine Julien ◽  
Eric Auclair ◽  
...  

2002 ◽  
Vol 17 (S2) ◽  
pp. S48
Author(s):  
Robyn R. M. Gershon ◽  
Kristine A. Qureshi ◽  
Stephen S. Morse ◽  
Marissa A. Berrera ◽  
Catherine B. Dela Cruz

Sign in / Sign up

Export Citation Format

Share Document