Abstract

Type 1 diabetes (T1D) arises secondary to immune-driven destruction of pancreatic β-cells and manifests as insulin-deficient hyperglycemia. We showed that oral vaccination with live attenuated Salmonella, which simultaneously delivers autoantigens and a TGFβ expression vector to immune cells in the gut mucosa, provides protection against the progression of T1D in non-obese diabetic (NOD) mice. In this study we employed the Sleeping Beauty (SB) transposon system that is composed of a transposase and transposon encoding the td-Tomato to express red fluorescent protein (RFP) to permanently mark the cells that take up the Salmonella vaccine. After animal vaccination, the transposon labeled-dendritic cells (DCs) with red fluorescence appeared throughout the secondary lymphoid tissues. Furthermore, Sleeping Beauty containing tgfβ1 gene (SB-tgfβ1) co-expressed TGFβ and RFP. The labeled DCs were detected predominantly in Peyer’s patches (PP) and mesenteric lymph nodes (MLN) and expressed CD103 surface marker. CD103+ DCs induced tolerogenic effects and gut homing. TGFβ significantly increased programmed death-ligand-1 (PDL-1 or CD274) expression in the DCs in the MLN and PP of treated mice. Also, TGFβ increased cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) levels in CD4+ cells in MLN and PP. Interestingly, DCs increased in all lymphatic organs of mice vaccinated with oral live Salmonella-based vaccine expressing preproinsulin (PPI), in combination with TGFβ, IL10, and subtherapeutic-doses of anti-CD3 mAb compared with vehicle-treated mice. These DCs are mostly tolerogenic in MLN and PP. Furthermore the DCs obtained from vaccine-treated but not vehicle-treated mice suppressed in vitro T cell proliferation. These data suggest that the MLN and the PP are a central hub for the beneficial anti-diabetic effects of an oral Salmonella-based vaccine prevention of diabetes in rodents.

Highlights

  • Type 1 diabetes (T1D) results from auto-reactive killing of the pancreatic islet insulin-producing β-cells [1]

  • The expression of TGFβ was detected in supernatant only after infection of macrophages with pSBbi-RP-TGFβ and pCMV(CAT)T7-SB100 (Figure 1B), implying that TGFβ was secreted after expression by host cells under the control of the CMV promoter

  • We found that CD4+CD25+ Treg cells from vehicle-treated mice efficiently suppressed proliferation and activation of responder T cells when combined with equal amount of dendritic cells (DCs) from vaccinated mice but did not alter proliferation when cultured with DCs from vehicletreated animals (Figure 7A)

Read more

Summary

Introduction

Type 1 diabetes (T1D) results from auto-reactive killing of the pancreatic islet insulin-producing β-cells [1]. Oral Vaccine for Type 1 Diabetes antigen-specific vaccines of live attenuated (non-pathogenic) Salmonella typhimurium diabetic autoantigens have been delivered to mice [6, 7]. Fusion of heterologous antigens to specific SPI2-T3SS proteins causes them to be presented to lymphocytes within the gut mucosa [12, 19, 20] This minimizes and/or bypasses intestinal lumen antigen expression and subsequent degradation of the same. A novel diabetes vaccine enabled direct expression of tolerogenic cytokines like TGFβ and IL10 and induced tolerance to diabetic auto-antigens [10, 23] These APCs process and produce the antigens which migrate to other organs in the gut and stimulate the immune cells [9, 24]

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