Elsevier

Microbial Pathogenesis

Volume 149, December 2020, 104565
Microbial Pathogenesis

Proteins involved in actin filament organization are key host factors for Japanese encephalitis virus life-cycle in human neuronal cells

https://doi.org/10.1016/j.micpath.2020.104565Get rights and content

Highlights

  • JEV can infect neuronal cells through clathrin-independent endocytosis.

  • Actin filament organization proteins are crucial for JEV infection in neuronal cells.

  • Small molecule inhibitors CK-548 and Wiskostatin inhibit JEV infection.

  • Proteins ATG12, BECN1, VAPA, VAPB and VCP are host-factors for JEV replication.

Abstract

Multiple membrane trafficking networks operate in the eukaryotic cell and are hijacked by viruses to establish infection. Recent studied have highlighted that viruses can exploit distinct pathways depending on the cell type. Japanese encephalitis virus (JEV), a neurotropic flavivirus, can infect neuronal cells through a clathrin-independent endocytic mechanism. To further characterize the membrane trafficking requirements for JEV infection of neuronal cells, we have performed a RNA interference-based study targeting 136 proteins in the human cell line IMR-32. Through quantitative RT-PCR and plaque assays we have validated that JEV infection in neuronal cells was independent of clathrin, and identified host-factors that were crucial for establishment of infection. Several of these proteins were involved in regulation of actin filament organization such as RHOA, RAC1, proteins of the ARP2/3 complex and N-WASP family, LIMK1, PAK1 and ROCK2. The small molecule inhibitors of ARP2/3 complex, CK-548 and of the N-WASP, Wiskostatin inhibited virus replication highlighting the important roles of these proteins in the virus life-cycle. We also identified ATG12, BECN1, VAPA, VAPB and VCP proteins as crucial host-factors for JEV replication across epithelial and neuronal cell lineages.

Introduction

Virus-host interaction involves a series of complex molecular and signaling events that are largely determined by the host cell lineage [[1], [2], [3], [4], [5]]. For several enveloped viruses, infection initiates with binding mediated by attachment factors and specific receptors, followed by internalization into one of the several endocytic portals operating at the plasma membrane [[6], [7], [8], [9], [10]]. Understanding the membrane trafficking requirements for pathogenic viruses is crucial for rational design of anti-virals [[11], [12], [13]].

Japanese encephalitis virus (JEV), a neurotropic flavivirus is a leading global cause of viral encephalitis with high morbidity and mortality [[14], [15], [16]]. Most flaviviruses adopt clathrin mediated endocytosis (CME) for establishing infection [8,[17], [18], [19]], however independent studies have now conclusively established that while JEV uses CME to enter epithelial cells and fibroblasts [[20], [21], [22], [23]], it infects neuronal cells through clathrin-independent endocytosis (CIE) [21,24,25]. The CIE of JEV in mouse neuronal cells was found to require dynamin, cholestrol, Rab5 and a dynamic actin network [21], while CIE in rat neuronal cells required dynamin and caveolin-1 [24]. RNA interference based studies have demonstrated that JEV replication in the human neuronal cell line SK-N-SH required caveolin-1, along with RHOA and RAC1 mediated actin cytoskeleton rearrangements [25]. Another recent study in human brain microvascular endothelial cells (HBMEC) has reconfirmed a role for CIE, with an essential role for Src and ezrin-mediated actin cytoskeleton polymerization for JEV entry [26].

Studies from our lab based on RNA interference screening of membrane trafficking genes in the epithelial cell line HeLa showed that JEV depends extensively on components of CME pathway for establishing infection, along with the members of the ARP2/3 complex [23]. Here we have utilized the same library to identify membrane trafficking host-factors essential for JEV replication in the human neuronal cell line IMR-32. The Rho family GTPAse RAC1 formed a central node with proteins of the two actin based processes playing a crucial role-the ARP2/3 complex and N-WASP family; and the PAK1-ROCK-2-LIMK1 signaling complex. We also identify other common host factors: ATG12, BECN1, VAPA, VAPB & VCP for JEV infection across epithelial and neuronal cell lineages.

Section snippets

Cells and virus

Human neuronal cell line IMR-32, and porcine stable kidney cell line- PS were obtained from National Centre for Cell Science (NCCS), Pune, India. SH-SY5Y and HeLa (CCL-2) cell lines were obtained from ATCC. IMR-32, SH-SY5Y and HeLa cell lines were cultured in Dulbecco's modified Eagle's medium (DMEM), and PS cells in minimum essential medium (MEM) supplemented with 10% fetal bovine serum (FBS) and 100 IU/ml Penicillin, 100 μg/ml Streptomycin and 2 mM l-Glutamine (PSG) solution. JEV strain

Establishment of siRNA screen in human neuronal IMR-32 cells

As a first step, we optimised the siRNA transfection conditions in IMR-32 cells to achieve maximum transfection efficiency. This was established by co-transfection of the non-targeting (NT) siRNA, or clathrin light chain (CLTB) siRNA with siGLO Green transfection indicator, which is a fluorescent oligonucleotide duplex that localizes to the nucleus (Fig. 1a). Fluorescence quantification of siGLO indicated that a transfection efficiency of 80–90% was achieved in IMR-32 cells (Fig. 1a).

We also

Discussion

Membrane trafficking networks are involved at multiple steps of the virus life-cycle ranging from entry to egress [6,9,54]. Most flaviviruses including JEV, have been shown to exploit a CME pathway to establish infection [8,[17], [18], [19]]. However, studies in the past ten years have shown that several viruses can opt for more than one route for endocytosis, depending on the cell type, which could be potentially caused by different receptor usage and/or activation of specific signalling

Funding information

This work was supported by DBT grant BT/MB/01/VIDRC/08 and by DBT intra-mural research funds to THSTI & RCB. RK was supported by ICMR-SRF fellowship, and was a PhD scholar with Symbiosis International (Deemed University), Symbiosis Knowledge Village, Gram Lavale, Mulshi Taluka, Pune, Maharashtra 412115, India. PS is supported by UGC-JRF fellowship.

Author statement

The authors declare that they have no known competing financial interest or personal relationship that influenced this study.

Declaration of competing interest

None.

Acknowledgements

We all thank all members of the Virology labs in THSTI & RCB.

References (74)

  • A.P.A. Ferreira et al.

    Mechanisms of carrier formation during clathrin-independent endocytosis

    Trends Cell Biol.

    (2018)
  • L. Galluzzi et al.

    Autophagy-independent functions of the autophagy machinery

    Cell

    (2019)
  • T.W. Koh et al.

    Synaptotagmin I, a Ca2+ sensor for neurotransmitter release

    Trends Neurosci.

    (2003)
  • T.H. Sollner

    Intracellular and viral membrane fusion: a uniting mechanism

    Curr. Opin. Cell Biol.

    (2004)
  • D. Panda

    Genome-wide RNAi screen identifies SEC61A and VCP as conserved regulators of Sindbis virus entry

    Cell Rep.

    (2013)
  • C.J. Merrifield

    Neural Wiskott Aldrich Syndrome Protein (N-WASP) and the Arp2/3 complex are recruited to sites of clathrin-mediated endocytosis in cultured fibroblasts

    Eur. J. Cell Biol.

    (2004)
  • T.D. Pollard et al.

    Structure and function of the Arp2/3 complex

    Curr. Opin. Struct. Biol.

    (2002)
  • J. Pizarro-Cerda

    The diverse family of arp2/3 complexes

    Trends Cell Biol.

    (2017)
  • K. Bosl

    Common nodes of virus-host interaction revealed through an integrated network analysis

    Front. Immunol.

    (2019)
  • Y.S. Ooi

    An RNA-centric dissection of host complexes controlling flavivirus infection

    Nat Microbiol

    (2019)
  • H. Ramage et al.

    Virus-host interactions: from unbiased genetic screens to function

    Annu Rev Virol

    (2015)
  • M. Laureti

    Flavivirus receptors: diversity, identity, and cell entry

    Front. Immunol.

    (2018)
  • M. Nain

    Japanese encephalitis virus invasion of cell: allies and alleys

    Rev. Med. Virol.

    (2016)
  • M. Mazzon et al.

    Targeting viral entry as a strategy for broad-spectrum antivirals

    F1000Res

    (2019)
  • Q.Y. Wang et al.

    Flavivirus entry inhibitors

    ACS Infect. Dis.

    (2015)
  • M.K. Zakaria et al.

    Cellular targets for the treatment of flavivirus infections

    Front Cell Infect Microbiol

    (2018)
  • A. Banerjee et al.

    Recent advances in understanding Japanese encephalitis

    F1000Res

    (2019)
  • T.C. Pierson et al.

    The continued threat of emerging flaviviruses

    Nat Microbiol

    (2020)
  • T.M. Quan

    Estimates of the global burden of Japanese encephalitis and the impact of vaccination from 2000-2015

    Elife

    (2020)
  • C. Cruz-Oliveira

    Receptors and routes of dengue virus entry into the host cells

    FEMS Microbiol. Rev.

    (2015)
  • B.A. Hackett et al.

    Flavivirus internalization is regulated by a size-dependent endocytic pathway

    Proc. Natl. Acad. Sci. U. S. A.

    (2018)
  • M. Nawa

    Interference in Japanese encephalitis virus infection of Vero cells by a cationic amphiphilic drug, chlorpromazine

    J. Gen. Virol.

    (2003)
  • M. Kalia

    Japanese encephalitis virus infects neuronal cells through a clathrin-independent endocytic mechanism

    J. Virol.

    (2013)
  • S. Yang

    Japanese encephalitis virus infects porcine kidney epithelial PK15 cells via clathrin- and cholesterol-dependent endocytosis

    Virol. J.

    (2013)
  • R. Khasa

    Membrane trafficking RNA interference screen identifies a crucial role of the clathrin endocytic pathway and ARP2/3 complex for Japanese encephalitis virus infection in HeLa cells

    J. Gen. Virol.

    (2019)
  • Y.Z. Zhu

    Japanese encephalitis virus enters rat neuroblastoma cells via a pH-dependent, dynamin and caveola-mediated endocytosis pathway

    J. Virol.

    (2012)
  • Q. Xu

    Caveolin-1-mediated Japanese encephalitis virus entry requires a two-step regulation of actin reorganization

    Future Microbiol.

    (2016)
  • Cited by (4)

    • Pathobiology of Japanese encephalitis virus infection

      2021, Molecular Aspects of Medicine
      Citation Excerpt :

      The deployment of a differential endocytic pathway in neuronal cells is likely to be governed by receptor usage. RNA interference-based screens have also identified several proteins involved in actin dynamics and other key host factors such as ezrin, platelet-derived growth factor receptor beta, and valosin-containing protein that are essential for virus replication (Khasa et al., 2019, 2020; Xu et al., 2016; Liu et al., 2020a; Sehrawat, 2021; Zhou et al., 2021). Similarly, CRISPR screening using a porcine sgRNA library also identified several critical host factors associated with JEV replication in porcine cells (Zhao et al., 2020).

    • Japanese Encephalitis Virus-Infected Cells

      2023, Subcellular Biochemistry
    1

    Present address: Department of Biological Science, Florida State University, Tallahassee, Florida, USA.

    View full text