Polyriboinosinic polyribocytidylic acid [poly(I:C)]/TLR3 signaling allows class I processing of exogenous protein and induction of HIV‐specific CD8+ cytotoxic T …

C Fujimoto, Y Nakagawa, K Ohara… - International …, 2004 - academic.oup.com
C Fujimoto, Y Nakagawa, K Ohara, H Takahashi
International immunology, 2004academic.oup.com
In the case of viral infection, various viral proteins and genetic components are disseminated
in the body. The former viral proteins may be captured by immature dendritic cells (DC) and
the latter genetic components may stimulate the antigen‐loading DC to maturate via specific
Toll‐like receptors (TLR), leading to the establishment of virus‐specific cellular immunity; in
particular, cytotoxic T lymphocytes (CTL) that control intracellular virions. Polyriboinosinic
polyribocytidylic acid [poly (I: C)], which might reflect a natural genetic product from a variety …
Abstract
In the case of viral infection, various viral proteins and genetic components are disseminated in the body. The former viral proteins may be captured by immature dendritic cells (DC) and the latter genetic components may stimulate the antigen‐loading DC to maturate via specific Toll‐like receptors (TLR), leading to the establishment of virus‐specific cellular immunity; in particular, cytotoxic T lymphocytes (CTL) that control intracellular virions. Polyriboinosinic polyribocytidylic acid [poly(I:C)], which might reflect a natural genetic product from a variety of viruses during replication, has recently been identified as one of the critical stimuli for TLR3. Based on these observations, we speculated that stimulation of TLR3 with poly(I:C) might drive the direction of acquired/adaptive immunity to the cellular arm. Indeed, when BALB/c mice were immunized with purified recombinant HIV‐1 envelope gp120 or influenza hemagglutinin (HA) protein together with poly(I:C), epitope‐specific CD8+ class I MHC molecule‐restricted CTL were primed from naive CD8+ T cells in vivo. In contrast, when the same proteins were immunized with lipopolysaccharide, a stimulant of TLR4, specific CTL were not primed at all. Moreover, we show here that immature DC could present processed antigen from captured purified protein in association with class I MHC molecules in the presence of poly(I:C), but not of LPS. These results indicate that we are able to manipulate the direction of acquired/adaptive effector immune responses using an appropriate stimuli and the findings presented in this paper will offer a new therapeutic strategy using poly(I:C) administration for priming antigen‐specific CD8+ CTL with purified viral protein in vivo.
Oxford University Press