Announcement

Collapse
No announcement yet.

The G protein-coupled receptor FFAR2 promotes internalization during influenza A virus entry

Collapse
X
 
  • Filter
  • Time
  • Show
Clear All
new posts

  • The G protein-coupled receptor FFAR2 promotes internalization during influenza A virus entry


    J Virol. 2019 Nov 6. pii: JVI.01707-19. doi: 10.1128/JVI.01707-19. [Epub ahead of print] The G protein-coupled receptor FFAR2 promotes internalization during influenza A virus entry.

    Wang G1, Jiang L1, Wang J1, Zhang J1, Kong F1, Li Q1, Yan Y1, Huang S1, Zhao Y1, Liang L1, Li J1, Sun N1, Hu Y1, Shi W1, Deng G1, Chen P1, Liu L1, Zeng X1, Tian G1, Bu Z1, Chen H1, Li C2.
    Author information

    1 State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China. 2 State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China lichengjun@caas.cn.

    Abstract

    Influenza A virus (IAV) co-opts numerous host factors to complete its replication cycle. Here, we identified free fatty acid receptor 2 (FFAR2) as a cofactor for IAV entry into host cells. We found that downregulation of FFAR2 or Ffar2 expression significantly reduced the replication of IAV in A549 or RAW 264.7 cells. The treatment of A549 cells with FFAR2 siRNA or the FFAR2 pathway agonists 4-CMTB and compound 58 [Cmp 58; (S)-2-(4-chlorophenyl)-3,3-dimethyl-N-(5-phenylthiazol-2-yl)butanamide], dramatically inhibited the nuclear accumulation of viral NP protein at early timepoints post infection, indicating that FFAR2 functions in the early stage of IAV replication cycle. FFAR2 downregulation had no effect on the expression of sialic acid (SA) receptors on the cell membrane, the attachment of IAV to the SA receptors, or the activity of the viral ribonucleoprotein (RNP) complex. Rather, the amount of internalized IAVs was significantly reduced in FFAR2 knocked-down, 4-CMTB-, or Cmp 58-treated A549 cells. Further studies showed that FFAR2 associated with β-arrestin1 and that β-arrestin1 interacted with the β2-subunit of the AP-2 complex (AP2B1), the essential adaptor of the clathrin-mediated endocytosis pathway. Notably, siRNA knockdown of either β-arrestin1 or AP2B1 dramatically impaired IAV replication, and AP2B1 knockdown or treatment with Barbadin, an inhibitor targeting the β-arrestin1/AP2B1 complex, remarkably decreased the amount of internalized IAVs. Moreover, we found that FFAR2 interacted with three GPCR kinases (i.e., GRK2, GRK5, and GRK6) whose downregulation inhibited IAV replication. Together, our findings demonstrate that the FFAR2 signaling cascade is important for the efficient endocytosis of IAV into host cells.IMPORTANCE To complete its replication cycle, IAV hijacks the host endocytosis machinery to invade cells. However, the underlying mechanisms of how IAV is internalized into host cells remain poorly understood, emphasizing the need to elucidate the role of host factors in IAV entry into cells. In this study, we identified FFAR2 as an important host factor for the efficient replication of both low pathogenic and highly pathogenic IAV. We revealed that FFAR2 facilitates the internalization of IAV into target cells during the early stage of infection. Upon further characterization of the role of FFAR2-associated proteins in virus replication, we found that the FFAR2-β-arrestin1-AP2B1 signaling cascade is important for the efficient endocytosis of IAV. Our findings thus further our understanding of the biological details of IAV entry into host cells and establish FFAR2 as a potential target for antiviral drug development.
    Copyright ? 2019 Wang et al.


    PMID: 31694949 DOI: 10.1128/JVI.01707-19
    Free full text

Working...
X