Multiple layers of CD80/86-dependent costimulatory activity regulate primary, memory, and secondary lymphocytic choriomeningitis virus-specific T cell immunity

J Eberlein, B Davenport, TT Nguyen, F Victorino… - Journal of …, 2012 - Am Soc Microbiol
J Eberlein, B Davenport, TT Nguyen, F Victorino, T Sparwasser, D Homann
Journal of virology, 2012Am Soc Microbiol
The lymphocytic choriomeningitis virus (LCMV) system constitutes one of the most widely
used models for the study of infectious disease and the regulation of virus-specific T cell
immunity. However, with respect to the activity of costimulatory and associated regulatory
pathways, LCMV-specific T cell responses have long been regarded as relatively
independent and thus distinct from the regulation of T cell immunity directed against many
other viral pathogens. Here, we have reevaluated the contribution of CD28-CD80/86 …
Abstract
The lymphocytic choriomeningitis virus (LCMV) system constitutes one of the most widely used models for the study of infectious disease and the regulation of virus-specific T cell immunity. However, with respect to the activity of costimulatory and associated regulatory pathways, LCMV-specific T cell responses have long been regarded as relatively independent and thus distinct from the regulation of T cell immunity directed against many other viral pathogens. Here, we have reevaluated the contribution of CD28-CD80/86 costimulation in the LCMV system by use of CD80/86-deficient mice, and our results demonstrate that a disruption of CD28-CD80/86 signaling compromises the magnitude, phenotype, and/or functionality of LCMV-specific CD8+ and/or CD4+ T cell populations in all stages of the T cell response. Notably, a profound inhibition of secondary T cell immunity in LCMV-immune CD80/86-deficient mice emerged as a composite of both defective memory T cell development and a specific requirement for CD80 but not CD86 in the recall response, while a related experimental scenario of CD28-dependent yet CD80/86-independent secondary CD8+ T cell immunity suggests the existence of a CD28 ligand other than CD80/86. Furthermore, we provide evidence that regulatory T cells (TREGs), the homeostasis of which is altered in CD80/86−/− mice, contribute to restrained LCMV-specific CD8+ T cell responses in the presence of CD80/86. Our observations can therefore provide a more coherent perspective on CD28-CD80/86 costimulation in antiviral T cell immunity that positions the LCMV system within a shared context of multiple defects that virus-specific T cells acquire in the absence of CD28-CD80/86 costimulation.
American Society for Microbiology