Regulation of hepatitis C virus infection by cellular retinoic acid binding proteins through the modulation of lipid droplet abundance

BR Bang, M Li, KN Tsai, H Aoyagi, SA Lee… - Journal of …, 2019 - Am Soc Microbiol
BR Bang, M Li, KN Tsai, H Aoyagi, SA Lee, K Machida, H Aizaki, JU Jung, JHJ Ou, T Saito
Journal of Virology, 2019Am Soc Microbiol
Retinoid (vitamin A) is an essential diet constituent that governs a broad range of biological
processes. Its biologically active metabolite, all-trans retinoic acid (ATRA), exhibits a potent
antiviral property by enhancing both innate and adaptive antiviral immunity against a variety
of viral pathogens, such as, but not limited to, HIV, respiratory syncytial virus (RSV), herpes
simplex virus (HSV), and measles. Even though the hepatocyte is highly enriched with
retinoid and its metabolite ATRA, it supports the establishment of efficient hepatitis C virus …
Abstract
Retinoid (vitamin A) is an essential diet constituent that governs a broad range of biological processes. Its biologically active metabolite, all-trans retinoic acid (ATRA), exhibits a potent antiviral property by enhancing both innate and adaptive antiviral immunity against a variety of viral pathogens, such as, but not limited to, HIV, respiratory syncytial virus (RSV), herpes simplex virus (HSV), and measles. Even though the hepatocyte is highly enriched with retinoid and its metabolite ATRA, it supports the establishment of efficient hepatitis C virus (HCV) replication. Here, we demonstrate the hepatocyte-specific cell-intrinsic mechanism by which ATRA exerts either a proviral or antiviral effect, depending on how it engages cellular retinoic acid binding proteins (CRABPs). We found that the engagement of CRABP1 by ATRA potently supported viral infection by promoting the accumulation of lipid droplets (LDs), which robustly enhanced the formation of a replication complex on the LD-associated endoplasmic reticulum (ER) membrane. In contrast, ATRA binding to CRABP2 potently inhibited HCV via suppression of LD accumulation. However, this antiviral effect of CRABP2 was abrogated due to the functional and quantitative predominance of CRABP1 in the hepatocytes. In summary, our study demonstrates that CRABPs serve as an on-off switch that modulates the efficiency of the HCV life cycle and elucidates how HCV evades the antiviral properties of ATRA via the exploitation of CRABP1 functionality.
IMPORTANCE ATRA, a biologically active metabolite of vitamin A, exerts pleiotropic biological effects, including the activation of both innate and adaptive immunity, thereby serving as a potent antimicrobial compound against numerous viral pathogens. Despite the enrichment of hepatocytes with vitamin A, HCV still establishes an efficient viral life cycle. Here, we discovered that the hepatocellular response to ATRA creates either a proviral or an antiviral environment depending on its engagement with CRABP1 or -2, respectively. CRABP1 supports the robust replication of HCV, while CRABP2 potently inhibits the efficiency of viral replication. Our biochemical, genetic, and microscopic analyses reveal that the pro- and antiviral effects of CRABPs are mediated by modulation of LD abundance, where HCV establishes the platform for viral replication and assembly on the LD-associated ER membrane. This study uncovered a cell-intrinsic mechanism by which HCV exploits the proviral function of CRABP1 to establish an efficient viral life cycle.
American Society for Microbiology