Expression of endoplasmic reticulum stress markers in the islets of patients with type 1 diabetes

Diabetologia ◽  
2012 ◽  
Vol 55 (9) ◽  
pp. 2417-2420 ◽  
Author(s):  
I. Marhfour ◽  
X. M. Lopez ◽  
D. Lefkaditis ◽  
I. Salmon ◽  
F. Allagnat ◽  
...  
Diabetologia ◽  
2013 ◽  
Vol 56 (12) ◽  
pp. 2638-2646 ◽  
Author(s):  
Chaoxing Yang ◽  
Philip diIorio ◽  
Agata Jurczyk ◽  
Bryan O’Sullivan-Murphy ◽  
Fumihiko Urano ◽  
...  

2015 ◽  
Vol 2015 ◽  
pp. 1-11 ◽  
Author(s):  
Antonia Diaz-Ganete ◽  
Gloria Baena-Nieto ◽  
Isabel M. Lomas-Romero ◽  
Jose Francisco Lopez-Acosta ◽  
Irene Cozar-Castellano ◽  
...  

Ghrelin is a peptidic hormone, which stimulates cell proliferation and inhibits apoptosis in several tissues, including pancreas. In preclinical stage of type 1 diabetes, proinflammatory cytokines generate a destructive environment forβ-cells known as insulitis, which results in loss ofβ-cell mass and impaired insulin secretion, leading to diabetes. Our aim was to demonstrate that ghrelin could preserveβ-cell viability, turnover rate, and insulin secretion acting as a counter balance of cytokines. In the present work we reproduced proinflammatory milieu found in insulitis stage by treating murine cell line INS-1E and rat islets with a cytokine cocktail including IL-1β, IFNγ, and TNFαand/or ghrelin. Several proteins involved in survival pathways (ERK 1/2 and Akt/PKB) and apoptosis (caspases and Bcl-2 protein family and endoplasmic reticulum stress markers) as well as insulin secretion were analyzed. Our results show that ghrelin alone has no remarkable effects onβ-cells in basal conditions, but interestingly it activates cell survival pathways, downregulates apoptotic mediators and endoplasmic reticulum stress, and restores insulin secretion in response to glucose when beta-cells are cytokine-exposed. These data suggest a potential role of ghrelin in preventing or slowing down the transition from a preclinical to clinically established diabetes by ameliorating the effects of insulitis onβ-cells.


Diabetes ◽  
2012 ◽  
Vol 61 (4) ◽  
pp. 818-827 ◽  
Author(s):  
S. A. Tersey ◽  
Y. Nishiki ◽  
A. T. Templin ◽  
S. M. Cabrera ◽  
N. D. Stull ◽  
...  

2020 ◽  
Vol 133 (1) ◽  
pp. 68-73
Author(s):  
Zhao-Hui Cao ◽  
Zhuan Wu ◽  
Cong Hu ◽  
Min Zhang ◽  
Wu-Zhou Wang ◽  
...  

2021 ◽  
Author(s):  
Xiaohui Zhang ◽  
Kelaier Yang ◽  
Jinyu Chi ◽  
Wenjia Chen ◽  
Xiao Ma ◽  
...  

Abstract Human recombinant relaxin-3 (H3 relaxin ),a small molecule peptide hormone, ameliorated myocardial injury after myocardial infarction or isoprenaline injection by inhibiting apoptosis and fibrosis. However, whether H3 relaxin protects vascular function in rats with type 1 diabetes and its mechanism are unknown. In type 1 diabetes rats model induced by streptozotocin (STZ), rats were subcutaneously injected H3 relaxin (2 µg/kg/d or 0.2 µg/kg/d) for 2 weeks. At 4 or 8 weeks after STZ injection, we detected the expression of fibrosis (type I and III collagen), ERS (endoplasmic reticulum stress) and NLRP3 inflammasome activation in the aortas and inflammation markers in the plasma from rats with diabetes. Compared with the diabetic rats, H3 relaxin treatment exhibited markedly decreased plasma oxidative stress markers (TNF-a and MDA) levels. The protein expression levels of type I and III collagen in the aortas were increased in rats with diabetes, inhibited by H3 relaxin. H3 relaxin treatment inhibited ERS (GRP78 and CHOP) and NLRP3 inflammasome activation in the aortas of diabetic rats. These results suggest that H3 relaxin inhibited fibrosis, ERS and inflammation activation in the aortas of type 1 diabetic rats.


2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Stéphane Demine ◽  
Andrea Alex Schiavo ◽  
Sandra Marín-Cañas ◽  
Piero Marchetti ◽  
Miriam Cnop ◽  
...  

Abstract Background Adult human pancreatic beta cells are the “gold standard” for studies on diabetes pathogenesis, but their use is limited by insufficient availability and variable quality. An important effort has recently taken place to differentiate beta cells from human induced pluripotent stem cells (iPSCs) and validate their use for diabetes research. We presently used a 7-stage protocol to generate beta cells from human iPSC and evaluated whether these cells are responsive to the pro-inflammatory cytokines (IFNγ, IL-1β, or IFNα) that play a role in type 1 diabetes. Methods The iPSC-derived islet-like cell clusters contained 40–50% beta and 10–15% alpha cells and expressed the receptors for IFNγ, IL-1β, or IFNα. Cells were exposed to either IFNγ (1000 U/mL) + IL-1β (50 U/mL) or IFNα alone (2000 U/mL) for 24/48 h. Apoptosis was quantified using Hoechst/propidium iodide staining or the RealTime Glo Apoptosis Kit (Promega). After treatment, CXCL10 secretion was quantified by ELISA. The expression of multiples genes (Ins, Gcg, Nkx2.2, Nkx6.1, Pdx1, Mafa, BiP, Chop, Atf3, CXCL10, CXCL9, CCL5, and HLA-ABC) was quantified by RT-qPCR. Phosphorylation state and total expression of STAT1/STAT2, as well as expression of PDL1 and of the ER chaperone BiP, were quantified by Western blotting. The co-localization of HLA-ABC or cleaved caspase-3 and Ins/Gcg expression was assessed by immunohistochemistry. The presence of HLA-ABC at the plasma membrane was measured by flow cytometry. Results IFNγ + IL-1β and IFNα induced apoptosis of the cells after 48 h of exposure. Cleaved caspase-3 co-localized mostly but not exclusively with Ins+ cells. Exposure to IFNγ + IL-1β induced a pro-inflammatory phenotype, including increased CXCL10, CXCL9, and CCL5 expression; CXCL10 secretion; and HLA-ABC expression. HLA overexpression was confirmed at the protein level by Western blotting and flow cytometry. Exposure to IFNγ + IL-1β (but not IFNα) also induced beta cell dedifferentiation and endoplasmic reticulum stress (increase in BiP, Chop, and Atf3 mRNA expression). Phosphorylation of STAT1 was stimulated already after 1 h by IFNγ + IL-1β and IFNα, while phosphorylation of STAT2 was only activated by IFNα at 1–4 h. PDL1 expression was increased by both IFNγ + IL-1β and IFNα. Conclusions Our data show that human iPSC-derived beta cells respond to pro-inflammatory cytokines IL-1β + IFNγ and IFNα, by activating the same pathogenic processes as adult human primary beta cells. These cells thus represent a valuable tool for future research on the pathogenesis of type 1 diabetes.


Diabetologia ◽  
2017 ◽  
Vol 60 (4) ◽  
pp. 656-667 ◽  
Author(s):  
Laura Marroqui ◽  
Reinaldo S. Dos Santos ◽  
Anne Op de beeck ◽  
Alexandra Coomans de Brachène ◽  
Lorella Marselli ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document