Abstract

Gut commensal bacteria are known to have a significant role in regulating the innate and adaptive immune homeostasis. Alterations in the intestinal microbial composition have been associated with several disease states, including autoimmune and inflammatory conditions. However, it is not entirely clear how commensal gut microbiota modulate and contribute to the systemic immunity, and whether circulating elements of the host immune system could regulate the microbiome. Thus, we have studied the diversity and abundance of specific taxons in the gut microbiota of inbred GalT-KO mice during 7 months of animal life by metagenetic high-throughput sequencing (16S rRNA gene, variable regions V3–V5). The repertoire of glycan-specific natural antibodies, obtained by printed glycan array technology, was then associated with the microbial diversity for each animal by metagenome-wide association studies (MWAS). Our data show that the orders clostridiales (most abundant), bacteriodales, lactobacillales, and deferribacterales may be associated with the development of the final repertoire of natural anti-glycan antibodies in GalT-KO mice. The main changes in microbiota diversity (month-2 and month-3) were related to important changes in levels and repertoire of natural anti-glycan antibodies in these mice. Additionally, significant positive and negative associations were found between the gut microbiota and the pattern of specific anti-glycan antibodies. Regarding individual features, the gut microbiota and the corresponding repertoire of natural anti-glycan antibodies showed differences among the examined animals. We also found redundancy in different taxa associated with the development of specific anti-glycan antibodies. Differences in microbial diversity did not, therefore, necessarily influence the overall functional output of the gut microbiome of GalT-KO mice. In summary, the repertoire of natural anti-carbohydrate antibodies may be partially determined by the continuous antigenic stimulation produced by the gut bacterial population of each GalT-KO mouse. Small differences in gut microbiota diversity could determine different repertoire and levels of natural anti-glycan antibodies and consequently might induce different immune responses to pathogens or other potential threats.

Highlights

  • Humans are colonized by trillions of microbial cells [1], the majority of this microbial ecosystem residing in the gut

  • The animal model used in this study spontaneously produced high blood levels of anti-α1-3 galactose (αGal) antibodies due to the inactivation of the enzyme α1,3-galactosyltransferase (Figure 1)

  • In the Printed Glycan Array (PGA) we considered values above 4,000 Relative Fluorescence Units (RFU) as a positive signal of antibody binding, which were expressed as the median ± IQR

Read more

Summary

Introduction

Humans are colonized by trillions of microbial cells [1], the majority of this microbial ecosystem residing in the gut. GM has a profound primary influence on human nutrition (digestion and absorption of nutrients), and metabolism, and seems to play a critical role in the development and function of the host immune system [5]. Alterations in composition and function of human GM have been associated with several pathologies, including metabolic disorders such as type-2 diabetes [6], obesity [7]; cardiovascular diseases [8]; autoimmune diseases such as inflammatory bowel disease [9], type-1 diabetes [10]; cancer [11], and diseases related to the central nervous system like Alzheimer’s and Parkinson’s diseases [12], and multiple sclerosis [13]. The interplay between the immune system and GM is very complex, and the underlying molecular mechanisms of host-microorganism interactions remain largely unknown [14]

Objectives
Results
Discussion
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