Cardiomyopathy and Diastolic Dysfunction in the Embryo and Neonate of a Type 1 Diabetic Mouse Model

2012 ◽  
Vol 20 (7) ◽  
pp. 781-790 ◽  
Author(s):  
Niamh Corrigan ◽  
Ann Treacy ◽  
Derek P. Brazil ◽  
Fionnuala M. McAuliffe
2016 ◽  
Vol 419 (1-2) ◽  
pp. 1-9 ◽  
Author(s):  
Zhe Hong ◽  
Zongyuan Hong ◽  
Denglong Wu ◽  
Hezhongrong Nie

2013 ◽  
Vol 190 (7) ◽  
pp. 3109-3120 ◽  
Author(s):  
Xiaotian Lin ◽  
Emma E. Hamilton-Williams ◽  
Daniel B. Rainbow ◽  
Kara M. Hunter ◽  
Yang D. Dai ◽  
...  

Cell Cycle ◽  
2020 ◽  
Vol 19 (23) ◽  
pp. 3329-3347
Author(s):  
Xiao-Tan Zhang ◽  
Guang Wang ◽  
Liu-Fang Ye ◽  
Yu Pu ◽  
Run-Tong Li ◽  
...  

PLoS ONE ◽  
2019 ◽  
Vol 14 (10) ◽  
pp. e0217728 ◽  
Author(s):  
Daniel R. Holohan ◽  
Frédéric Van Gool ◽  
Jeffrey A. Bluestone

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

2019 ◽  
Vol 21 (1) ◽  
pp. 71-77 ◽  
Author(s):  
Janice M. Nieves-Bonilla ◽  
Badr Kiaf ◽  
Cornelia Schuster ◽  
Stephan Kissler

2019 ◽  
Author(s):  
Daniel R. Holohan ◽  
Frédéric Van Gool ◽  
Jeffrey A. Bluestone

AbstractRegulatory T cells (Tregs) are an immunosuppressive population that are identified based on the stable expression of the fate-determining transcription factor forkhead box P3 (Foxp3). Tregs can be divided into distinct subsets based on whether they developed in the thymus (tTregs) or in the periphery (pTregs). Whether there are unique functional roles that distinguish pTregs and tTregs remains largely unclear. To elucidate these functions, efforts have been made to specifically identify and modify individual Treg subsets. Deletion of the conserved non-coding sequence (CNS)1 in the Foxp3 locus leads to selective impairment of pTreg generation without disrupting tTreg generation in the C57BL/6J background. Using CRISPR-Cas9 genome editing technology, we removed the Foxp3 CNS1 region in the non-obese diabetic (NOD) mouse model of spontaneous type 1 diabetes mellitus (T1D) to determine if pTregs contribute to autoimmune regulation. Deletion of CNS1 impaired in vitro induction of Foxp3 in naïve NOD CD4+ T cells, but it did not alter Tregs in most lymphoid and non-lymphoid tissues analyzed except for the large intestine lamina propria, where a small but significant decrease in RORγt+ Tregs and corresponding increase in Helios+ Tregs was observed in NOD CNS1−/− mice. CNS1 deletion also did not alter the development of T1D or glucose tolerance despite increased pancreatic insulitis in pre-diabetic female NOD CNS1−/− mice. CNS1 Furthermore, the proportions of autoreactive Tregs and conventional T cells (Tconvs) within pancreatic islets were unchanged. These results suggest that pTregs dependent on the Foxp3 CNS1 region are not the dominant regulatory population controlling T1D in the NOD mouse model.


Sign in / Sign up

Export Citation Format

Share Document