While there is growing evidence for tumor-associated antigens in renal cell carcinoma (RCC), most spontaneous immune responses to the disease are ineffective at limiting tumor progression. One mechanism by which tumors promote their own progressive growth is by inducing or sensitizing T cells to apoptosis. We recently determined that the ability of several RCC cell lines to mediate significant levels of apoptosis in co-cultured T cells is largely abrogated when the co-incubations are performed either in the presence of neutralizing anti-TNF antibodies, or with ganglioside-depleted tumor cells. This suggestion of a synergistic effect between tumor-derived gangliosides and TNF for inducing T cell apoptosis is corroborated by studies with purified sources of TNF and the RCC-overexpressed ganglioside GM1, which demonstrate similar cooperativity for killing T cells when used together in vitro. Our previous work indicated that select gangliosides can inhibit NFkappaB activation in T cells, thereby sensitizing the lymphocytes to apoptosis by preventing their synthesis of NFkappaB-dependent, anti-apoptotic proteins. The mechanism by which gangliosides cause that defect, and the nature of the second signal actually inducing apoptosis, however, have remained undefined. Preliminary data now suggest that RCC-derived gangliosides and TNF synergize to induce T cell apoptosis by two different mechanisms. We hypothesize that tumor-derived gangliosides disrupt lipid rafts on T cell membranes, preventing effective NFkappaB activation through TNFR1, and hence rendering apoptosis the default pathway induced upon TNF binding. We also have evidence that TNF enhances the effectiveness with which RCC gangliosides induce proapoptotic effects on the mitochondrion, accelerating the onset and intensity of the ROS and MPT mediated by gangliosides alone. The experiments outlined in this proposal will both assess the importance of a TNF/ganglioside synergy to RCC-induced T cell apoptosis, and test our hypothesis that both of the proposed, synergistic mechanisms may be contributing to the T cell dysfunction in RCC.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA111917-04
Application #
7333208
Study Section
Cancer Immunopathology and Immunotherapy Study Section (CII)
Program Officer
Mccarthy, Susan A
Project Start
2005-01-15
Project End
2008-12-31
Budget Start
2008-01-01
Budget End
2008-12-31
Support Year
4
Fiscal Year
2008
Total Cost
$257,865
Indirect Cost
Name
Cleveland Clinic Lerner
Department
Other Basic Sciences
Type
Schools of Medicine
DUNS #
135781701
City
Cleveland
State
OH
Country
United States
Zip Code
44195
Sa, Gaurisankar; Das, Tanya; Moon, Christina et al. (2009) GD3, an overexpressed tumor-derived ganglioside, mediates the apoptosis of activated but not resting T cells. Cancer Res 69:3095-104
Biswas, Soumika; Biswas, Kaushik; Richmond, Amy et al. (2009) Elevated levels of select gangliosides in T cells from renal cell carcinoma patients is associated with T cell dysfunction. J Immunol 183:5050-8
Das, Tanya; Sa, Gaurisankar; Paszkiewicz-Kozik, Ewa et al. (2008) Renal cell carcinoma tumors induce T cell apoptosis through receptor-dependent and receptor-independent pathways. J Immunol 180:4687-96
Das, Tanya; Sa, Gaurisankar; Hilston, Cynthia et al. (2008) GM1 and tumor necrosis factor-alpha, overexpressed in renal cell carcinoma, synergize to induce T-cell apoptosis. Cancer Res 68:2014-23
Raval, Gira; Biswas, Soumika; Rayman, Patricia et al. (2007) TNF-alpha induction of GM2 expression on renal cell carcinomas promotes T cell dysfunction. J Immunol 178:6642-52