Prevention of autoimmune diabetes in NOD mice by troglitazone is associated with modulation of ICAM-1 expression on pancreatic islet cells and IFN-γ expression in splenic T cells

2003 ◽  
Vol 304 (2) ◽  
pp. 378-384 ◽  
Author(s):  
Petra Augstein ◽  
Annemarie Dunger ◽  
Peter Heinke ◽  
Gerhild Wachlin ◽  
Sabine Berg ◽  
...  
Diabetologia ◽  
2003 ◽  
Vol 46 (2) ◽  
pp. 255-266 ◽  
Author(s):  
A. K. Cardozo ◽  
P. Proost ◽  
C. Gysemans ◽  
M.-C. Chen ◽  
C. Mathieu ◽  
...  

2020 ◽  
Vol 117 (49) ◽  
pp. 31319-31330
Author(s):  
Hao Hu ◽  
Pavel N. Zakharov ◽  
Orion J. Peterson ◽  
Emil R. Unanue

Autoimmune diabetes is one of the complications resulting from checkpoint blockade immunotherapy in cancer patients, yet the underlying mechanisms for such an adverse effect are not well understood. Leveraging the diabetes-susceptible nonobese diabetic (NOD) mouse model, we phenocopy the diabetes progression induced by programmed death 1 (PD-1)/PD-L1 blockade and identify a cascade of highly interdependent cellular interactions involving diabetogenic CD4 and CD8 T cells and macrophages. We demonstrate that exhausted CD8 T cells are the major cells that respond to PD-1 blockade producing high levels of IFN-γ. Most importantly, the activated T cells lead to the recruitment of monocyte-derived macrophages that become highly activated when responding to IFN-γ. These macrophages acquire cytocidal activity against β-cells via nitric oxide and induce autoimmune diabetes. Collectively, the data in this study reveal a critical role of macrophages in the PD-1 blockade-induced diabetogenesis, providing new insights for the understanding of checkpoint blockade immunotherapy in cancer and infectious diseases.


2017 ◽  
Vol 114 (48) ◽  
pp. E10418-E10427 ◽  
Author(s):  
Javier A. Carrero ◽  
Derrick P. McCarthy ◽  
Stephen T. Ferris ◽  
Xiaoxiao Wan ◽  
Hao Hu ◽  
...  

Treatment of C57BL/6 or NOD mice with a monoclonal antibody to the CSF-1 receptor resulted in depletion of the resident macrophages of pancreatic islets of Langerhans that lasted for several weeks. Depletion of macrophages in C57BL/6 mice did not affect multiple parameters of islet function, including glucose response, insulin content, and transcriptional profile. In NOD mice depleted of islet-resident macrophages starting at 3 wk of age, several changes occurred: (i) the early entrance of CD4 T cells and dendritic cells into pancreatic islets was reduced, (ii) presentation of insulin epitopes by dispersed islet cells to T cells was impaired, and (iii) the development of autoimmune diabetes was significantly reduced. Treatment of NOD mice starting at 10 wk of age, when the autoimmune process has progressed, also significantly reduced the incidence of diabetes. Despite the absence of diabetes, NOD mice treated with anti–CSF-1 receptor starting at 3 or 10 wk of age still contained variably elevated leukocytic infiltrates in their islets when examined at 20–40 wk of age. Diabetes occurred in the anti–CSF-1 receptor protected mice after treatment with a blocking antibody directed against PD-1. We conclude that treatment of NOD mice with an antibody against CSF-1 receptor reduced diabetes incidence and led to the development of a regulatory pathway that controlled autoimmune progression.


2020 ◽  
Vol 11 ◽  
Author(s):  
Tingting Ge ◽  
Gaurang Jhala ◽  
Stacey Fynch ◽  
Satoru Akazawa ◽  
Sara Litwak ◽  
...  

Cytokines that signal through the JAK-STAT pathway, such as interferon-γ (IFN-γ) and common γ chain cytokines, contribute to the destruction of insulin-secreting β cells by CD8+ T cells in type 1 diabetes (T1D). We previously showed that JAK1/JAK2 inhibitors reversed autoimmune insulitis in non-obese diabetic (NOD) mice and also blocked IFN-γ mediated MHC class I upregulation on β cells. Blocking interferons on their own does not prevent diabetes in knockout NOD mice, so we tested whether JAK inhibitor action on signaling downstream of common γ chain cytokines, including IL-2, IL-7 IL-15, and IL-21, may also affect the progression of diabetes in NOD mice. Common γ chain cytokines activate JAK1 and JAK3 to regulate T cell proliferation. We used a JAK1-selective inhibitor, ABT 317, to better understand the specific role of JAK1 signaling in autoimmune diabetes. ABT 317 reduced IL-21, IL-2, IL-15 and IL-7 signaling in T cells and IFN-γ signaling in β cells, but ABT 317 did not affect GM-CSF signaling in granulocytes. When given in vivo to NOD mice, ABT 317 reduced CD8+ T cell proliferation as well as the number of KLRG+ effector and CD44hiCD62Llo effector memory CD8+ T cells in spleen. ABT 317 also prevented MHC class I upregulation on β cells. Newly diagnosed diabetes was reversed in 94% NOD mice treated twice daily with ABT 317 while still on treatment at 40 days and 44% remained normoglycemic after a further 60 days from discontinuing the drug. Our results indicate that ABT 317 blocks common γ chain cytokines in lymphocytes and interferons in lymphocytes and β cells and are thus more effective against diabetes pathogenesis than IFN-γ receptor deficiency alone. Our studies suggest use of this class of drug for the treatment of type 1 diabetes.


2021 ◽  
Author(s):  
Vasu Patel ◽  
Arathi Jayaraman ◽  
Sundararajan Jayaraman

The histone modifier Trichostatin A (TSA) ameliorated diabetes and repressed IFN-γ and IL-17A expression in prediabetic female NOD mice. Purified CD4+ cells could be polarized ex vivo into Th1 and Th17 subsets, which comparably transferred diabetes into NOD.scid mice. Polarized Th1 cells were devoid of IL-17A-producing cells and did not transdifferentiate into Th17 cells in an immunodeficient environment. However, Th17 cells had contaminant Th1 cells, which expressed IFN-γ upon adoptive transfer into lymphopenic recipients. Notably, TSA treatment abrogated the transfer of diabetes by CD4+ T-cells cultured under Th1 or Th17 polarizing conditions accompanied by the absence of Ifng and Il17a expression in NOD.scid recipients. Significantly, the histone modifier restored the ability of CD4+ but not CD8+ T-cells to undergo CD3-mediated apoptosis ex vivo in a caspase-dependent manner. Thus, the histone modifier afforded protection against autoimmune diabetes by negative regulation of signature lymphokines and restitution of self-tolerance in CD4+ T cells.


Cell Cycle ◽  
2010 ◽  
Vol 9 (15) ◽  
pp. 3199-3201 ◽  
Author(s):  
Qing-Sheng Mi ◽  
Hong-Zhi He ◽  
Zheng Dong ◽  
Carlos Isales ◽  
Li Zhou

Diabetes ◽  
1980 ◽  
Vol 29 (6) ◽  
pp. 497-500 ◽  
Author(s):  
P. Meda ◽  
E. L. Hooghe-Peters ◽  
L. Orci

Diabetes ◽  
1988 ◽  
Vol 37 (8) ◽  
pp. 1123-1128
Author(s):  
N. Welsh ◽  
A. Hallberg ◽  
S. Sandler ◽  
C. Hellerstrom

Sign in / Sign up

Export Citation Format

Share Document