scholarly journals Generation and Characterization of Alloantigen-Specific Regulatory T Cells For Clinical Transplant Tolerance

2018 ◽  
Vol 8 (1) ◽  
Author(s):  
James M. Mathew ◽  
Jessica H. Voss ◽  
Scott T. McEwen ◽  
Iwona Konieczna ◽  
Arjun Chakraborty ◽  
...  
2016 ◽  
Vol 77 ◽  
pp. 68
Author(s):  
James M. Mathew ◽  
Jessica L. Heinrichs ◽  
Scott T. McEwen ◽  
Iwona M. Konieczna ◽  
Xuemei Huang ◽  
...  

2011 ◽  
Vol 16 (6) ◽  
pp. 606-613 ◽  
Author(s):  
Julien Zuber ◽  
Olivier Hermine ◽  
Lucienne Chatenoud ◽  
Christophe Legendre

2014 ◽  
Vol 28 (S1) ◽  
Author(s):  
Gabriella Tedeschi ◽  
Elisa Maffioli ◽  
Armando Negri ◽  
Andrea Romagnani ◽  
Fabio Grassi

2021 ◽  
Author(s):  
Yannick D. Muller ◽  
Leonardo M.R. Ferreira ◽  
Emilie Ronin ◽  
Patrick Ho ◽  
Vinh Nguyen ◽  
...  

Infusion of regulatory T cells (Tregs) engineered with a chimeric antigen receptor (CAR) targeting donor-derived human leukocyte antigen (HLA) is a promising strategy to promote transplant tolerance. Here, we describe an anti-HLA-A2 CAR (A2-CAR) generated by grafting the complementarity-determining regions (CDRs) of a human monoclonal anti-HLA-A2 antibody into the framework regions of the Herceptin 4D5 single-chain variable fragment and fusing it with a CD28-zeta signaling domain. The CDR-grafted A2-CAR maintained the specificity of the original antibody. We then generated HLA-A2 mono-specific human CAR Tregs either by deleting the endogenous T-cell receptor (TCR) via CRISPR/Cas9 and introducing the A2-CAR using lentiviral transduction or by directly integrating the CAR construct into the TCR alpha constant locus using homology-directed repair. These A2-CAR+TCRdeficient human Tregs maintained both Treg phenotype and function in vitro. Moreover, they selectively accumulated in HLA-A2-expressing islets transplanted from either HLA-A2 transgenic mice or deceased human donors. A2-CAR+TCRdeficient Tregs did not impair the function of these HLA-A2+ islets, whereas similarly engineered A2-CAR+TCRdeficientCD4+ conventional T cells rejected the islets in less than 2 weeks. A2-CAR+TCRdeficient Tregs delayed graft-versus-host disease only in the presence of HLA-A2, expressed either by co-transferred peripheral blood mononuclear cells or by the recipient mice. Altogether, we demonstrate that genome-engineered mono-antigen-specific A2-CAR Tregs localize to HLA-A2-expressing grafts and exhibit antigen-dependent in vivo suppression, independent of TCR expression. These approaches may be applied towards developing precision Treg cell therapies for transplant tolerance.


2018 ◽  
Vol 4 (4) ◽  
pp. 205-213 ◽  
Author(s):  
Eman Shaban ◽  
George Bayliss ◽  
Deepak K. Malhotra ◽  
Douglas Shemin ◽  
Li Juan Wang ◽  
...  

2018 ◽  
Vol 102 ◽  
pp. S109
Author(s):  
Juewan Kim ◽  
Zhilian Yue ◽  
Xiao Liu ◽  
Christopher Hope ◽  
Darling Rojas-Canales ◽  
...  

2020 ◽  
Vol 204 (4) ◽  
pp. 858-867
Author(s):  
Annette Ko ◽  
Masashi Watanabe ◽  
Thomas Nguyen ◽  
Alvin Shi ◽  
Achouak Achour ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document