5
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      TLR5-deficiency controls dendritic cell subset development in an autoimmune diabetes-susceptible model

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Introduction

          The incidence of the autoimmune disease, type 1 diabetes (T1D), has been increasing worldwide and recent studies have shown that the gut microbiota are associated with modulating susceptibility to T1D. Toll-like receptor 5 (TLR5) recognizes bacterial flagellin and is widely expressed on many cells, including dendritic cells (DCs), which are potent antigen-presenting cells (APCs). TLR5 modulates susceptibility to obesity and alters metabolism through gut microbiota; however, little is known about the role TLR5 plays in autoimmunity, especially in T1D.

          Methods

          To fill this knowledge gap, we generated a TLR5-deficient non-obese diabetic (NOD) mouse, an animal model of human T1D, for study.

          Results

          We found that TLR5-deficiency led to a reduction in CD11c + DC development in utero, prior to microbial colonization, which was maintained into adulthood. This was associated with a bias in the DC populations expressing CD103, with or without CD8α co-expression, and hyper-secretion of different cytokines, both in vitro (after stimulation) and directly ex vivo. We also found that TLR5-deficient DCs were able to promote polyclonal and islet antigen-specific CD4 + T cell proliferation and proinflammatory cytokine secretion. Interestingly, only older TLR5-deficient NOD mice had a greater risk of developing spontaneous T1D compared to wild-type mice.

          Discussion

          In summary, our data show that TLR5 modulates DC development and enhances cytokine secretion and diabetogenic CD4+ T cell responses. Further investigation into the role of TLR5 in DC development and autoimmune diabetes may give additional insights into the pathogenesis of Type 1 diabetes.

          Related collections

          Most cited references63

          • Record: found
          • Abstract: found
          • Article: not found

          Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity.

          Although in vitro observations suggest that cross-presentation of antigens is mediated primarily by CD8alpha+ dendritic cells, in vivo analysis has been hampered by the lack of systems that selectively eliminate this cell lineage. We show that deletion of the transcription factor Batf3 ablated development of CD8alpha+ dendritic cells, allowing us to examine their role in immunity in vivo. Dendritic cells from Batf3-/- mice were defective in cross-presentation, and Batf3-/- mice lacked virus-specific CD8+ T cell responses to West Nile virus. Importantly, rejection of highly immunogenic syngeneic tumors was impaired in Batf3-/- mice. These results suggest an important role for CD8alpha+ dendritic cells and cross-presentation in responses to viruses and in tumor rejection.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Temporal development of the gut microbiome in early childhood from the TEDDY study

            The development of the microbiome from infancy to childhood is dependent on a range of factors, with microbial–immune crosstalk during this time thought to be involved in the pathobiology of later life diseases1–9 such as persistent islet autoimmunity and type 1 diabetes10–12. However, to our knowledge, no studies have performed extensive characterization of the microbiome in early life in a large, multi-centre population. Here we analyse longitudinal stool samples from 903 children between 3 and 46 months of age by 16S rRNA gene sequencing (n = 12,005) and metagenomic sequencing (n = 10,867), as part of the The Environmental Determinants of Diabetes in the Young (TEDDY) study. We show that the developing gut microbiome undergoes three distinct phases of microbiome progression: a developmental phase (months 3–14), a transitional phase (months 15–30), and a stable phase (months 31–46). Receipt of breast milk, either exclusive or partial, was the most significant factor associated with the microbiome structure. Breastfeeding was associated with higher levels of Bifidobacterium species (B. breve and B. bifidum), and the cessation of breast milk resulted in faster maturation of the gut microbiome, as marked by the phylum Firmicutes. Birth mode was also significantly associated with the microbiome during the developmental phase, driven by higher levels of Bacteroides species (particularly B. fragilis) in infants delivered vaginally. Bacteroides was also associated with increased gut diversity and faster maturation, regardless of the birth mode. Environmental factors including geographical location and household exposures (such as siblings and furry pets) also represented important covariates. A nested case–control analysis revealed subtle associations between microbial taxonomy and the development of islet autoimmunity or type 1 diabetes. These data determine the structural and functional assembly of the microbiome in early life and provide a foundation for targeted mechanistic investigation into the consequences of microbial–immune crosstalk for long-term health.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids.

              GPR41 and GPR43 are related members of a homologous family of orphan G protein-coupled receptors that are tandemly encoded at a single chromosomal locus in both humans and mice. We identified the acetate anion as an agonist of human GPR43 during routine ligand bank screening in yeast. This activity was confirmed after transient transfection of GPR43 into mammalian cells using Ca(2+) mobilization and [(35)S]guanosine 5'-O-(3-thiotriphosphate) binding assays and by coexpression with GIRK G protein-regulated potassium channels in Xenopus laevis oocytes. Other short chain carboxylic acid anions such as formate, propionate, butyrate, and pentanoate also had agonist activity. GPR41 is related to GPR43 (52% similarity; 43% identity) and was activated by similar ligands but with differing specificity for carbon chain length, with pentanoate being the most potent agonist. A third family member, GPR42, is most likely a recent gene duplication of GPR41 and may be a pseudogene. GPR41 was expressed primarily in adipose tissue, whereas the highest levels of GPR43 were found in immune cells. The identity of the cognate physiological ligands for these receptors is not clear, although propionate is known to occur in vivo at high concentrations under certain pathophysiological conditions.
                Bookmark

                Author and article information

                Contributors
                URI : https://loop.frontiersin.org/people/549102Role: Role: Role: Role: Role: Role: Role: Role: Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/2161444Role: Role: Role: Role: Role: Role:
                Role: Role: Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/626993Role: Role: Role:
                URI : https://loop.frontiersin.org/people/549028Role: Role: Role: Role: Role: Role: Role: Role: Role:
                Journal
                Front Immunol
                Front Immunol
                Front. Immunol.
                Frontiers in Immunology
                Frontiers Media S.A.
                1664-3224
                28 February 2024
                2024
                : 15
                : 1333967
                Affiliations
                [1] 1 Section of Endocrinology, School of Medicine, Yale University , New Haven, CT, United States
                [2] 2 Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University , Cardiff, United Kingdom
                Author notes

                Edited by: Salvatore Oliva, Sapienza University of Rome, Italy

                Reviewed by: Bin Bao, Boston Children’s Hospital and Harvard Medical School, United States

                Myung-Shik Lee, Yonsei University College of Medicine, Republic of Korea

                Bin Zhao, Central South University, China

                *Correspondence: James Alexander Pearson, pearsonj1@ 123456cardiff.ac.uk ; Li Wen, li.wen@ 123456yale.edu
                Article
                10.3389/fimmu.2024.1333967
                10935730
                38482010
                a4f1c465-4081-4201-8103-d2b231b6f19c
                Copyright © 2024 Pearson, Hu, Peng, Wong and Wen

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 06 November 2023
                : 06 February 2024
                Page count
                Figures: 6, Tables: 0, Equations: 0, References: 63, Pages: 13, Words: 6740
                Funding
                Funded by: National Institutes of Health , doi 10.13039/100000002;
                Award ID: HD097808, DK12689, DK130318
                Funded by: Diabetes Research Connection , doi 10.13039/100019970;
                Award ID: 2019-14
                Funded by: JDRF , doi 10.13039/100022690;
                Award ID: PDF-2016-197
                Funded by: Medical Research Council , doi 10.13039/501100000265;
                Award ID: MR/T010525/1
                The author(s) declare financial support was received for the research, authorship, and/or publication of this article. This work was supported by grants to LW from NIH (HD097808, DK12689 and DK130318) and a research grant from Diabetes Research Connection (2019–14), and to JAP, a JDRF Postdoctoral Research Fellowship (PDF-2016-197) and a Medical Research Council Career Development Award (MR/T010525/1).
                Categories
                Immunology
                Original Research
                Custom metadata
                Mucosal Immunity

                Immunology
                tlr5,nod mice,dendritic cells,type 1 diabetes,microbiota
                Immunology
                tlr5, nod mice, dendritic cells, type 1 diabetes, microbiota

                Comments

                Comment on this article