Abstract

Tight junction barrier is critical to intestinal homeostasis. Applying antibiotics to treat infections is common in clinical practice, which may affect intestinal microbiota. Intestinal microbiota dysbiosis is involved in the occurrence of some gastrointestinal diseases. Therefore, this study was aimed to investigate the influence of antibiotics on intestinal tight junction barrier and the possible underlying mechanisms. Healthy adult female C57BL/6 mice were treated with a broad-spectrum antibiotic cocktail for 14 days. 16S rDNA Illumina sequencing and headspace gas chromatography-mass spectrometry (HS-GC/MS) were respectively used to analyze microbial community and to detect short-chain fatty acids (SCFAs) contents. In vivo intestinal paracellular permeability to fluorescein isothiocyanate-dextran (FITC-dextran) was measured. Protein expression was determined by immunoblotting. Immunofluoresence was applied to observe the distributions of ZO-1, LC3B and ASC. Antibiotics remarkably altered intestinal microbiota composition in healthy mice, accompanying reduced SCFAs’ concentrations. In addition, the intestinal tight junction barrier was disrupted by antibiotic treatment, as evidenced by increased intestinal paracellular permeability to FITC-dextran, decreased tight junction protein expressions, and disrupted ZO-1 morphology. Furthermore, NLRP3 inflammasome and autophagy were activated by antibiotic treatment. In conclusion, intestinal epithelial tight junction barrier dysfunction induced by antibiotics is associated with intestinal microbiota dysbiosis, activated NLRP3 inflammasome and autophagy in mice.

Highlights

  • Antibiotics alters the diversity and composition of intestinal microbiota To figure out the alterations of intestinal microbiota after antibiotic treatment, we evaluated the composition and diversity through an Illumina high-throughput sequencing technique

  • We show that the potential biomarkers after antibiotics treatment might be Klebsiella, Parasutterella, Morganella, unclassified_f_Enterobacteriaceae, Ureaplasma and unclassified_f_Ruminococcaceae, majority of which belong to Proteobateria phylum

  • Antibiotics increase relative abundance of species whose functions are associated with the transport and metabolism of amino acids and carbohydrates, but reduce the species whose functions are related to cell membrane biogenesis, defense mechanisms, replication, recombination and repair

Read more

Summary

Introduction

Antibiotics, microbiota and barrier dysfunction resulting in intestinal homeostasis [1,2]. Disrupted intestinal epithelial tight junction barrier is compromised in a variety of diseases, such as inflammatory bowel disease, intestinal ischemia/reperfusion injury, shock, and severe burns [1,2,3,4,5,6]. It has been reported that intestinal microbiota contributes to intestinal epithelial barrier improvement, immune system development, nutrients absorption and pathogens colonization restriction [8]. The intestinal microbiota can be altered by many factors and diseases, including stress, intestinal ischemia/reperfusion, infection, dietary changes, and antibiotics [9,10,11,12], resulting in the imbalance of intestinal homeostasis

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call