scholarly journals Hyperglycemia-Induced Proliferation of Adult Human Beta Cells Engrafted Into Spontaneously Diabetic Immunodeficient NOD-Rag1null IL2rγnull Ins2Akita Mice

Pancreas ◽  
2011 ◽  
Vol 40 (7) ◽  
pp. 1147-1149 ◽  
Author(s):  
Philip diIorio ◽  
Agata Jurczyk ◽  
Chaoxing Yang ◽  
Waldemar J. Racki ◽  
Michael A. Brehm ◽  
...  
PLoS ONE ◽  
2012 ◽  
Vol 7 (4) ◽  
pp. e35801 ◽  
Author(s):  
Sabine Rutti ◽  
Nadine S. Sauter ◽  
Karim Bouzakri ◽  
Richard Prazak ◽  
Philippe A. Halban ◽  
...  

2019 ◽  
Vol 29 (3) ◽  
pp. 638-652.e5 ◽  
Author(s):  
Peng Wang ◽  
Esra Karakose ◽  
Hongtao Liu ◽  
Ethan Swartz ◽  
Courtney Ackeifi ◽  
...  

Diabetes ◽  
1998 ◽  
Vol 47 (1) ◽  
pp. 134-137 ◽  
Author(s):  
V. H. Lefebvre ◽  
T. Otonkoski ◽  
J. Ustinov ◽  
M. A. Huotari ◽  
D. G. Pipeleers ◽  
...  

Cell Reports ◽  
2016 ◽  
Vol 17 (12) ◽  
pp. 3281-3291 ◽  
Author(s):  
Joana Almaça ◽  
Judith Molina ◽  
Danusa Menegaz ◽  
Alexey N. Pronin ◽  
Alejandro Tamayo ◽  
...  

2018 ◽  
Vol 8 (1) ◽  
Author(s):  
Maria J. L. Kracht ◽  
Eelco J. P. de Koning ◽  
Rob C. Hoeben ◽  
Bart O. Roep ◽  
Arnaud Zaldumbide

Diabetologia ◽  
2000 ◽  
Vol 43 (4) ◽  
pp. 465-473 ◽  
Author(s):  
H. Rochlitz ◽  
A. Voigt ◽  
B. Lankat-Buttgereit ◽  
B. G�ke ◽  
H. Heimberg ◽  
...  

Cells ◽  
2020 ◽  
Vol 9 (1) ◽  
pp. 168 ◽  
Author(s):  
Anne-Françoise Close ◽  
Nidheesh Dadheech ◽  
Hélène Lemieux ◽  
Qian Wang ◽  
Jean Buteau

Nor1, the third member of the Nr4a subfamily of nuclear receptor, is garnering increased interest in view of its role in the regulation of glucose homeostasis. Our previous study highlighted a proapoptotic role of Nor1 in pancreatic beta cells and showed that Nor1 expression was increased in islets isolated from type 2 diabetic individuals, suggesting that Nor1 could mediate the deterioration of islet function in type 2 diabetes. However, the mechanism remains incompletely understood. We herein investigated the subcellular localization of Nor1 in INS832/13 cells and dispersed human beta cells. We also examined the consequences of Nor1 overexpression on mitochondrial function and morphology. Our results show that, surprisingly, Nor1 is mostly cytoplasmic in beta cells and undergoes mitochondrial translocation upon activation by proinflammatory cytokines. Mitochondrial localization of Nor1 reduced glucose oxidation, lowered ATP production rates, and inhibited glucose-stimulated insulin secretion. Western blot and microscopy images revealed that Nor1 could provoke mitochondrial fragmentation via mitophagy. Our study unveils a new mode of action for Nor1, which affects beta-cell viability and function by disrupting mitochondrial networks.


Diabetologia ◽  
2019 ◽  
Vol 63 (2) ◽  
pp. 395-409 ◽  
Author(s):  
Masaya Oshima ◽  
Séverine Pechberty ◽  
Lara Bellini ◽  
Sven O. Göpel ◽  
Mélanie Campana ◽  
...  

Abstract Aims/hypothesis During the onset of type 2 diabetes, excessive dietary intake of saturated NEFA and fructose lead to impaired insulin production and secretion by insulin-producing pancreatic beta cells. The majority of data on the deleterious effects of lipids on functional beta cell mass were obtained either in vivo in rodent models or in vitro using rodent islets and beta cell lines. Translating data from rodent to human beta cells remains challenging. Here, we used the human beta cell line EndoC-βH1 and analysed its sensitivity to a lipotoxic and glucolipotoxic (high palmitate with or without high glucose) insult, as a way to model human beta cells in a type 2 diabetes environment. Methods EndoC-βH1 cells were exposed to palmitate after knockdown of genes related to saturated NEFA metabolism. We analysed whether and how palmitate induces apoptosis, stress and inflammation and modulates beta cell identity. Results EndoC-βH1 cells were insensitive to the deleterious effects of saturated NEFA (palmitate and stearate) unless stearoyl CoA desaturase (SCD) was silenced. SCD was abundantly expressed in EndoC-βH1 cells, as well as in human islets and human induced pluripotent stem cell-derived beta cells. SCD silencing induced markers of inflammation and endoplasmic reticulum stress and also IAPP mRNA. Treatment with the SCD products oleate or palmitoleate reversed inflammation and endoplasmic reticulum stress. Upon SCD knockdown, palmitate induced expression of dedifferentiation markers such as SOX9, MYC and HES1. Interestingly, SCD knockdown by itself disrupted beta cell identity with a decrease in mature beta cell markers INS, MAFA and SLC30A8 and decreased insulin content and glucose-stimulated insulin secretion. Conclusions/interpretation The present study delineates an important role for SCD in the protection against lipotoxicity and in the maintenance of human beta cell identity. Data availability Microarray data and all experimental details that support the findings of this study have been deposited in in the GEO database with the GSE130208 accession code.


2020 ◽  
Vol 44 (7) ◽  
pp. S32-S33
Author(s):  
Su Wang ◽  
Stephane Flibotte ◽  
Joan Camunas-Soler ◽  
Patrick E. Macdonald ◽  
James Johnson

Sign in / Sign up

Export Citation Format

Share Document