Faculty Opinions recommendation of Protective antifungal memory CD8(+) T cells are maintained in the absence of CD4(+) T cell help and cognate antigen in mice.

Author(s):  
Sarah Gaffen ◽  
Nydiaris Hernandez-Santos
2012 ◽  
Vol 122 (3) ◽  
pp. 987-999 ◽  
Author(s):  
Som G. Nanjappa ◽  
Erika Heninger ◽  
Marcel Wüthrich ◽  
Thomas Sullivan ◽  
Bruce Klein

2010 ◽  
Vol 185 (6) ◽  
pp. 3436-3444 ◽  
Author(s):  
Daniel K. Choo ◽  
Kaja Murali-Krishna ◽  
Rustom Anita ◽  
Rafi Ahmed

Immunity ◽  
2014 ◽  
Vol 41 (4) ◽  
pp. 633-645 ◽  
Author(s):  
Brian J. Laidlaw ◽  
Nianzhi Zhang ◽  
Heather D. Marshall ◽  
Mathew M. Staron ◽  
Tianxia Guan ◽  
...  

Blood ◽  
2007 ◽  
Vol 109 (12) ◽  
pp. 5318-5326 ◽  
Author(s):  
Sandra Hervas-Stubbs ◽  
Aurélie Olivier ◽  
Florence Boisgerault ◽  
Nathalie Thieblemont ◽  
Claude Leclerc

Abstract We investigated whether Toll-like receptor ligands (TLR-Ls) can bypass the requirement for CD4+ T-cell help in the induction of fully efficient memory CD8+ T cells (cytotoxic T lymphocytes [CTLs]). “Helpless” CTLs were induced by a synthetic CD8+ T-cell epitope administered with TLR3-L and TLR9-L, but not with TLR2/6-L, TLR4-L, or TLR7-L. The up-regulation of MHC-I and costimulatory molecules by dendritic cells following TLR stimulation was not sufficient for the priming of “helpless” CTLs, which depended essentially on the induction of a strong IFN-α/β response. The “helpless” CTLs induced by TLR-Ls differentiated into fully functional memory CTLs able to proliferate as well as their “helped” counterparts upon challenge, in the absence of CD4+ T-cell help.


2006 ◽  
Vol 176 (4) ◽  
pp. 2316-2323 ◽  
Author(s):  
Nick D. Jones ◽  
Manuela Carvalho-Gaspar ◽  
Shiqiao Luo ◽  
Matthew O. Brook ◽  
Laurent Martin ◽  
...  

2012 ◽  
Vol 3 (1) ◽  
Author(s):  
Sonia Feau ◽  
Zacarias Garcia ◽  
Ramon Arens ◽  
Hideo Yagita ◽  
Jannie Borst ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3175-3175
Author(s):  
Sanju Jalla ◽  
Erin McCadden ◽  
Jie Wang ◽  
Ephraim J. Fuchs ◽  
Katharine A. Whartenby

Abstract Since CD4+ T cell help has been proposed to be required for maintaining the activity of tumor-specific CD8+ T cells, tolerance in tumor-specific CD4+ T cells may seriously impair the efficacy of therapeutic tumor vaccines. To overcome this problem, we devised a strategy to “engineer” CD4+ T cell help by treating tumor-bearing animals with nonmyeloablative conditioning and transplantation of autologous hematopoietic stem cells (HSCs) that have been genetically modified, via lentiviral transduction, to express an antigen containing “foreign” CD4+ T cell epitopes. After hematopoietic reconstitution, animals received the combination of an autologous tumor cell vaccine and an infusion of primed CD4+ T cells specific for the expressed epitopes. Using influenza hemagglutinin (HA) as the model antigen, we first confirmed that transplantation of HA-transduced HSCs led to efficient expression of HA by antigen-presenting cells, as demonstrated by the clonal expansion of adoptively transferred, HA-specific CD4+ transgenic T cells in mice receiving HA-transduced HSCs but not in mice receiving nerve growth factor receptor (NGFR) gene-transduced HSCs. Next, BALB/c mice harboring 13 day old, metastatic 4T1 mammary cancer were treated with removal of the primary, nonmyeloablative conditioning and transplantation of HA-transduced syngeneic HSCs, and following hematopoietic reconstitution, with concomitant autologous tumor cell vaccination and adoptive transfer of in vitro activated, HA-specific transgenic CD4+ T cells. This therapy was successful in curing the majority of tumor bearing mice, and was superior to the same therapy given to mice transplanted with NGFR-transduced stem cells. Finally, we found that the anti-tumor effect of vaccination plus exogenous T cell help was abolished by the adoptive transfer of either CD4+ or CD8+ T cells from tumor-bearing mice, suggesting that tumor-bearing mice contain both potential effectors and suppressors of anti-tumor immunity, the latter of which are abolished by the non-myeloablative conditioning. These results highlight the importance of CD4+ T cell help in the induction of therapeutic anti-tumor immunity.


Sign in / Sign up

Export Citation Format

Share Document