Your browser doesn't support javascript.
loading
Maternal milk and fecal microbes guide the spatiotemporal development of mucosa-associated microbiota and barrier function in the porcine neonatal gut.
Liu, Hongbin; Zeng, Xiangfang; Zhang, Guolong; Hou, Chengli; Li, Ning; Yu, Haitao; Shang, Lijun; Zhang, Xiaoya; Trevisi, Paolo; Yang, Feiyun; Liu, Zuohua; Qiao, Shiyan.
Afiliação
  • Liu H; State Key Laboratory of Animal Nutrition and Beijing Key Laboratory of Bio-Feed Additives, China Agricultural University, Beijing, China.
  • Zeng X; Present Address: Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
  • Zhang G; State Key Laboratory of Animal Nutrition and Beijing Key Laboratory of Bio-Feed Additives, China Agricultural University, Beijing, China.
  • Hou C; Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, USA.
  • Li N; Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China.
  • Yu H; State Key Laboratory of Animal Nutrition and Beijing Key Laboratory of Bio-Feed Additives, China Agricultural University, Beijing, China.
  • Shang L; State Key Laboratory of Animal Nutrition and Beijing Key Laboratory of Bio-Feed Additives, China Agricultural University, Beijing, China.
  • Zhang X; State Key Laboratory of Animal Nutrition and Beijing Key Laboratory of Bio-Feed Additives, China Agricultural University, Beijing, China.
  • Trevisi P; State Key Laboratory of Animal Nutrition and Beijing Key Laboratory of Bio-Feed Additives, China Agricultural University, Beijing, China.
  • Yang F; Department of Agricultural and Food Science, University of Bologna, Bologna, Italy.
  • Liu Z; Chongqing Academy of Animal Science, Chongqing, China.
  • Qiao S; Chongqing Academy of Animal Science, Chongqing, China.
BMC Biol ; 17(1): 106, 2019 12 18.
Article em En | MEDLINE | ID: mdl-31852478
ABSTRACT

BACKGROUND:

The early-life microbiota exerts a profound and lifelong impact on host health. Longitudinal studies in humans have been informative but are mostly based on the analysis of fecal samples and cannot shed direct light on the early development of mucosa-associated intestinal microbiota and its impact on GI function. Using piglets as a model for human infants, we assess here the succession of mucosa-associated microbiota across the intestinal tract in the first 35 days after birth.

RESULTS:

Although sharing a similar composition and predicted functional profile at birth, the mucosa-associated microbiome in the small intestine (jejunum and ileum) remained relatively stable, while that of the large intestine (cecum and colon) quickly expanded and diversified by day 35. Among detected microbial sources (milk, vagina, areolar skin, and feces of sows, farrowing crate, and incubator), maternal milk microbes were primarily responsible for the colonization of the small intestine, contributing approximately 90% bacteria throughout the first 35 days of the neonatal life. Although maternal milk microbes contributed greater than 90% bacteria to the large intestinal microbiota of neonates upon birth, their presence gradually diminished, and they were replaced by maternal fecal microbes by day 35. We found strong correlations between the relative abundance of specific mucosa-associated microbes, particularly those vertically transmitted from the mother, and the expression levels of multiple intestinal immune and barrier function genes in different segments of the intestinal tract.

CONCLUSION:

We revealed spatially specific trajectories of microbial colonization of the intestinal mucosa in the small and large intestines, which can be primarily attributed to the colonization by vertically transmitted maternal milk and intestinal microbes. Additionally, these maternal microbes may be involved in the establishment of intestinal immune and barrier functions in neonates. Our findings strengthen the notion that studying fecal samples alone is insufficient to fully understand the co-development of the intestinal microbiota and immune system and suggest the possibility of improving neonatal health through the manipulation of maternal microbiota.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Sus scrofa / Leite / Microbioma Gastrointestinal / Mucosa Intestinal / Intestinos Tipo de estudo: Observational_studies / Prognostic_studies / Risk_factors_studies Limite: Animals Idioma: En Revista: BMC Biol Assunto da revista: BIOLOGIA Ano de publicação: 2019 Tipo de documento: Article País de afiliação: China

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Sus scrofa / Leite / Microbioma Gastrointestinal / Mucosa Intestinal / Intestinos Tipo de estudo: Observational_studies / Prognostic_studies / Risk_factors_studies Limite: Animals Idioma: En Revista: BMC Biol Assunto da revista: BIOLOGIA Ano de publicação: 2019 Tipo de documento: Article País de afiliação: China