• JoomlaWorks Simple Image Rotator
  • JoomlaWorks Simple Image Rotator
  • JoomlaWorks Simple Image Rotator
  • JoomlaWorks Simple Image Rotator
  • JoomlaWorks Simple Image Rotator
  • JoomlaWorks Simple Image Rotator
  • JoomlaWorks Simple Image Rotator
  • JoomlaWorks Simple Image Rotator
  • JoomlaWorks Simple Image Rotator
  • JoomlaWorks Simple Image Rotator
 
  Bookmark and Share
 
 
Master's Dissertation
DOI
https://doi.org/10.11606/D.17.2019.tde-15102018-110809
Document
Author
Full name
Jefferson Antonio Leite
E-mail
Institute/School/College
Knowledge Area
Date of Defense
Published
Ribeirão Preto, 2018
Supervisor
Committee
Sartori, Daniela Carlos (President)
Pontillo, Alessandra
Faria, Ana Maria Caetano de
Zamboni, Dario Simões
Title in Portuguese
A ativação do receptor AIM2 na mucosa intestinal confere proteção ao diabetes tipo 1 experimental
Keywords in Portuguese
AIM2 ; Diabetes tipo 1 ; Microbiota ; Th1 ; Resposta imune inata
Abstract in Portuguese
O diabetes tipo 1 (DM1) é uma doença autoimune caracterizada pela destruição das células ? presentes nas ilhotas pancreáticas por linfócitos T autorreativos, especialmente Th1 e Th17, levando o indivíduo a um estado de hiperglicemia. Embora existam diversos estudos que abordam a resposta imune adaptativa no contexto do DM1, poucos trabalhos tentaram elucidar o papel da resposta imune inata no desenvolvimento da doença. Neste contexto, observamos que camundongos WT pré-diabéticos possuem um aumento significativo na expressão gênica e proteica do receptor AIM2 e de moléculas relacionadas à sua via de ativação e sinalização (Caspase-1, IL-1? e IL-18) nos linfonodos pancreáticos (LNPs) e no íleo. Posteriormente, foi verificado que camundongos deficientes do receptor AIM2 tornaram-se mais suscetíveis ao DM1, comprovado por elevados níveis de glicose sanguínea e menor produção de insulina em relação aos animais selvagens (WT) após a administração com estreptozotocina (STZ). Tal suscetibilidade está relacionada a um processo de disbiose e aumento da translocação de bactérias da microbiota intestinal para os LNPs de camundongos AIM2-/-. De maneira interessante, o inflamassoma AIM2 foi ativado apenas na presença de DNA fecal de animais diabéticos, que possui uma microbiota em disbiose, uma vez que resultou na produção significativa da citocina IL-1?. Também foi constatado que a ativação do receptor AIM2 na mucosa intestinal regulou a expressão gênica e proteica de proteínas de junção celular, peptídeos antimicrobianos e mucinas, como forma de minimizar a translocação de bactérias da microbiota para os LNPs. Adicionalmente, foi visto que a ativação do receptor AIM2 contribui para a indução de células Th17 intestinais, para a migração de neutrófilos no intestino, assim como para a expressão das citocinas IL-23, IL-17 e IL-22 no íleo. Por fim, mostramos que o receptor AIM2 modulou negativamente a ativação de células dendríticas expressando TLR4 e TLR9, que correlacionou com o aumento de células Tc1 patogênicas nos LNPs. De forma geral, nossos resultados demonstram que a ativação do receptor AIM2 na mucosa intestinal desempenha um importante papel em controlar a homeostase da microbiota intestinal, manter a integridade da barreira intestinal, e consequentemente.
Title in English
The activation of AIM2 receptor in the intestinal mucosal protects against experimental type 1 diabetes
Keywords in English
AIM2 ; Gut microbiota ; Innate immune response ; Th17 ; Type 1 diabetes
Abstract in English
Type 1 diabetes (T1D) is an autoimmune disease characterized by the destruction of ? cells present in the pancreatic islets by autoreactive T lymphocytes, especially Th1 and Th17, leading to a state of hyperglycemia. There are many studies that address the role of adaptive immune response, so only some studies have attempted to elucidate the role of the innate immune response in the context of T1D. In this regard, we observed that pre-diabetic WT mice have a significant increase in the gene and protein expression of the AIM2 receptor and in molecules related to its activation and signaling pathways (Caspase-1, IL- 1? and IL-18) in the pancreatic lymph nodes (PLNs) and in the ileum. Subsequently, it was verified that AIM2 receptor deficient mice became more susceptible to T1D, as proved by blood glucose levels and lower insulin production compared to wild-type mice (WT) after administration of streptozotocin (STZ). This susceptibility was related to a process of dysbiosis and increased translocation of bacteria from gut microbiota to PLNs in AIM2-/- mice. Interestingly, the AIM2 inflammasome was activated in the presence of fecal DNA from diabetic mice, which has a gut microbiota in dysbiosis, since resulted in significant production of IL-1?. It was found that activation of the AIM2 receptor in the intestinal mucosa regulated the gene and protein expression of tightjunction proteins, antimicrobial peptides and mucins in order to minimizing a bacterial translocation of the microbiota to the PLNs. In addition, it was seen that activation of the AIM2 receptor contributes to induction of intestinal Th17 cells, to neutrophil migration in the intestine, as well as for expression of IL-23, IL-17 and IL-22 cytokines in the ileum. Finally, we show that the AIM2 receptor negatively modulated the activation of dendritic cells expressing TLR4 and TLR9, which correlated with the increase of pathogenic Tc1 cells in the PLNs. In general, the results demonstrate that activation of the AIM2 receptor in the intestinal mucosa plays an important role in controlling the composition of gut microbiota homeostasis, maintaining the intestinal barrier function, and consequently reducing the bacterial translocation to the PLNs, conferring a protective effect to the immunopathogeny against to DM1.
 
WARNING - Viewing this document is conditioned on your acceptance of the following terms of use:
This document is only for private use for research and teaching activities. Reproduction for commercial use is forbidden. This rights cover the whole data about this document as well as its contents. Any uses or copies of this document in whole or in part must include the author's name.
Jefferson.pdf (4.48 Mbytes)
Publishing Date
2019-02-18
 
WARNING: Learn what derived works are clicking here.
All rights of the thesis/dissertation are from the authors
CeTI-SC/STI
Digital Library of Theses and Dissertations of USP. Copyright © 2001-2024. All rights reserved.