Elsevier

Antiviral Research

Volume 150, February 2018, Pages 217-225
Antiviral Research

Erythrosin B is a potent and broad-spectrum orthosteric inhibitor of the flavivirus NS2B-NS3 protease

https://doi.org/10.1016/j.antiviral.2017.12.018Get rights and content

Highlights

  • We identified erythrosin B as a potent inhibitor for flavivirus protease.

  • Erythrosin B inhibited the DENV2 protease via a non-competitive mechanism.

  • Erythrosin B interferes the interactions between viral NS3 and its co-factor NS2B.

  • Erythrosin B is a potent inhibitor for Zika virus and Dengue virus.

Abstract

Many flaviviruses, such as Zika virus (ZIKV), Dengue virus (DENV1-4) and yellow fever virus (YFV), are significant human pathogens. Infection with ZIKV, an emerging mosquito-borne flavivirus, is associated with increased risk of microcephaly in newborns and Guillain-Barré syndrome and other complications in adults. Currently, specific therapy does not exist for any flavivirus infections. In this study, we found that erythrosin B, an FDA-approved food additive, is a potent inhibitor for flaviviruses, including ZIKV and DENV2. Erythrosin B was found to inhibit the DENV2 and ZIKV NS2B-NS3 proteases with IC50 in low micromolar range, via a non-competitive mechanism. Erythrosin B can significantly reduce titers of representative flaviviruses, DENV2, ZIKV, YFV, JEV, and WNV, with micromolar potency and with excellent cytotoxicity profile. Erythrosin B can also inhibit ZIKV replication in ZIKV-relevant human placental and neural progenitor cells. As a pregnancy category B food additive, erythrosin B may represent a promising and easily developed therapy for management of infections by ZIKV and other flaviviruses.

Introduction

The genus Flavivirus contains more than 70 viruses, many of which cause serious human diseases. The four serotypes of Dengue virus (DENV1-4), yellow fever virus (YFV), West Nile virus (WNV), Zika virus (ZIKV), St. Louis encephalitis virus (SLEV), Japanese encephalitis virus (JEV), Powassan virus (POWV), and tick-borne encephalitis virus (TBEV) are categorized as global emerging pathogens. Due to global travel, ZIKV outbreaks recently occurred worldwide, and was imported to many new territories including the UK, Canada, and the US (Attar, 2016, Bogoch et al., 2016, Chen, 2016, Korhonen et al., 2016). ZIKV transmission has been further increased by transfer between humans through sexual activities and blood transfusions (Foy et al., 2011, Musso et al., 2014, Musso et al., 2015, Patino-Barbosa et al., 2015). Importantly, ZIKV infection increases the risk of microcephaly in babies born to infected mothers, and may cause Guillain-Barré syndrome and other complications in adults (Calvet et al., 2016, Martines et al., 2016, Rodriguez-Morales, 2016, Thomas et al., 2016). Published data in mice and humans have demonstrated that ZIKV infects the central nervous system during fetal development, including (but not limited to) regions like the cortex, thalamus, and hypothalamus, as well as neurogenic regions including the subventricular and subgranular zones, with fetal infection occurring through placenta cells (Cugola et al., 2016, Lancaster et al., 2013, Tabata et al., 2016, Tang et al., 2016). Infection with ZIKV results in a phenotypic and genetically measured infection in stem cell and neural progenitor cells with cortical thickness, cell apoptosis, and most critically, a phenotype similar to microcephaly (Cugola et al., 2016, Lancaster et al., 2013, Li et al., 2016, Tang et al., 2016).

The flavivirus genomic RNA is single-stranded with positive polarity. The viral genome is approximately 11 kb in length, composed of a 5′ untranslated region (UTR), a single long open reading frame (ORF), and a 3’ UTR. The single ORF encodes a large precursor polyprotein (PP) that requires post-translational processing by host and viral proteases (Chambers et al., 1990). These processes lead to mature viral proteins, including three structural proteins (capsid, pre-membrane or membrane, and envelope), and seven non-structural (NS) proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5). Among the viral proteins, NS3 is the second largest protein encoded by flavivirus, and has multi-enzymatic functions, including protease, RNA triphosphatase, nucleoside triphosphatase, and helicase activities (Brecher et al., 2013, Lim et al., 2013, Luo et al., 2015, Sampath and Padmanabhan, 2009). The viral protease is encoded by the N-terminal region (∼180 amino acids (aa)) of the NS3 protein. The viral NS2B protein is a membrane-associated protein, with a central hydrophobic core region (∼40 aa) as an essential co-factor for the NS3 protease function. As an essential enzyme, the flavivirus NS2B-NS3 protease is highly conserved (Brecher et al., 2013, Brecher et al., 2017, Chambers et al., 1991, Falgout et al., 1991).

In a recent study, we developed a split luciferase complementation (SLC)-based high-throughput screening assay to identify inhibitors blocking the essential interactions between the DENV2 protease NS3 and its co-factor NS2B (Li et al., 2017a). Pilot screening of the NCATS Chemical Genomic Center (NCGC) Pharmaceutical Collection compound library resulted in a few priority “hits” as potent orthosteric inhibitors capable of abolishing NS2B-NS3 interactions (Li et al., 2017a).

In this study, we explored additional hit compounds that were not characterized by previous study. Among these uncharacterized compounds, erythrosin B (EB), an FDA-approved food additive, was also found to act as a potent inhibitor for the flavivirus NS2B-NS3 protease. Erythrosin B moderately inhibited the interactions between the DENV2 NS2B and NS3 with IC50-SLC of 15 μM, and significantly inhibited the DENV2 and ZIKV protease activities with IC50-pro in low micromolar range via a non-competitive mechanism (IC50: half maximal inhibitory concentration, defined as the compound concentration required to inhibit protease activity (IC50-pro) or SLC (IC50-SLC) by 50%). Results from cell-based assays indicated that EB significantly reduced titers of DENV2, ZIKV, WNV, YFV, and JEV with EC50 in low micromolar potency (EC50: half maximal effective concentration, defined as the compound concentration required to reduce virus titer by 50%). In contrast, significantly higher concentrations of EB (>150 μM) will be required to cause death of human cells, including human placental epithelial cells and neural progenitor cells relevant to ZIKV pathogenesis. As an FDA pregnancy category B food additive, EB may represent a promising and easily developed therapy for management of infections by DENV, ZIKV and other flaviviruses.

Section snippets

Compounds

Erythrosin B was procured from Sigma-Aldrich.

Cloning, expression and purification

All clones were generated as described previously (Li et al., 2017a). The MBP-NS3 mutants were generated using the Strategene QuickChange II mutagenesis strategy with primer pairs shown in Supplemental Table S1. All proteins were expressed in Escherichia coli strain Rosetta 2(DE3) (EMD Biosciences) and purified through a nickel-nitrilotriacetic acid (NTA) column (Qiagen) or Glutathione sepharose 4B (GE HealthCare), followed by a gel filtration 16/60

Erythrosin B abolishes interactions between NS2B and NS3 of DENV2

Previously, we developed an SLC-based NS2B-NS3 interaction assay by generating GST-CLuc398-NS3 and His-NLuc416-NS2B49-66 fusion proteins in which the C-terminal fragment (CLuc398) of firefly luciferase (FLuc, aa 398–550) was fused between the GST-tag and viral NS3, and the N-terminal fragment (NLuc416) of FLuc (aa 1–416) was fused between the His-tag and viral NS2B peptide 49–66 (Li et al., 2017a). Interactions between the NS2B peptide 49–66 and NS3 result in complementation of NLuc and CLuc to

Discussion

Since the outbreak of WNV in the New York City in 1999, the flavivirus protease enzyme has been an encouraging drug target. Recent study shows that inhibitors preventing the NS2B-NS3 interaction provide a promising approach for therapeutic development (Li et al., 2017a).

In this study, we identified erythrosin B as a candidate orthosteric inhibitor blocking interactions between flavivirus NS2B and NS3. Erythrosin B, also known as Red No. 3, erythrosine, or food additive E127, is an organoiodine

Acknowledgement

We thank J. Tang at the Wadsworth Center Tissue Culture Core facility for help with cell culture and K. McClive-Reed for helpful suggestions. We also thank other core facilities at the Wadsworth Center, including the Applied Genomic Technologies Core for DNA sequencing, the Advanced Light Microscopy Core for immunofluorescence imaging, and the Tissue Culture Core for media. M.B. was partially supported by the NIH Biodefense and Emerging Infectious Disease training grant AI055429. This work was

References (45)

  • A. Sampath et al.

    Molecular targets for flavivirus drug discovery

    Antivir. Res.

    (2009)
  • T. Tabata et al.

    Zika virus targets different primary human placental cells, suggesting two routes for vertical transmission

    Cell Host Microbe

    (2016)
  • T. Tanaka

    Reproductive and neurobehavioural toxicity study of erythrosine administered to mice in the diet

    Food Chem. Toxicol.

    (2001)
  • H. Tang et al.

    Zika virus infects human cortical neural progenitors and attenuates their growth

    Cell Stem Cell

    (2016)
  • (ANS), E.P.o.F.A.a.N.S.a.t.F

    Scientific Opinion on the re-evaluation of Erythrosine (E 127) as a food additive

    EFSA J.

    (2011)
  • N. Attar

    ZIKA virus circulates in new regions

    Nat. Rev. Microbiol.

    (2016)
  • M. Brecher et al.

    Novel broad spectrum inhibitors targeting the flavivirus methyltransferase

    PLos One

    (2015)
  • M. Brecher et al.

    A conformational switch high-throughput screening assay and allosteric inhibition of the flavivirus NS2B-NS3 protease

    PLoS Pathog.

    (2017)
  • M. Brecher et al.

    The flavivirus protease as a target for drug discovery

    Virol. Sin.

    (2013)
  • S.M. Chambers et al.

    Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling

    Nat. Biotechnol.

    (2009)
  • T.J. Chambers et al.

    Processing of the yellow fever virus nonstructural polyprotein: a catalytically active NS3 proteinase domain and NS2B are required for cleavages at dibasic sites

    J. Virol.

    (1991)
  • T.J. Chambers et al.

    Flavivirus genome organization, expression, and replication

    Annu. Rev. Microbiol.

    (1990)
  • Cited by (58)

    • Allosteric quinoxaline-based inhibitors of the flavivirus NS2B/NS3 protease

      2023, Bioorganic Chemistry
      Citation Excerpt :

      Structural and binding mode characterizations have proven challenging for non-competitive inhibitors of flaviviral proteases. The non-competitive inhibitors previously reported were suggested to bind in the allosteric site or the NS2B binding site [24,25,40], but reliable cocrystal structures or direct binding affinity measurements have been elusive. In addition to the relatively modest potencies of those inhibitors [24–27], experimental characterization is challenged by the extremely dynamic nature of the protease and especially the NS2B cofactor.

    View all citing articles on Scopus
    View full text