Elsevier

Virus Research

Volume 244, 15 January 2018, Pages 64-70
Virus Research

Comparative analysis of different cell systems for Zika virus (ZIKV) propagation and evaluation of anti-ZIKV compounds in vitro

https://doi.org/10.1016/j.virusres.2017.11.003Get rights and content

Highlights

  • ZIKV replicates in human, mosquito and monkey cell lines.

  • Huh7 cells yield maximal production of infectious ZIKV.

  • Human A549 are effective as the reference nonhuman VERO cells in plaque assay test.

  • ZIKV RNA and PFU titers best correlated in Huh7-producing, A549-readout cells.

  • Comparable drug EC50 obtained by qRT-PCR and plaque assay in the Huh7/A549 system.

Abstract

A strong correlation between Zika virus (ZIKV) infection and severe neurological disease in newborns and occasionally adults has emerged in the Brazilian outbreak. Efficient human cell-based assays are required to test candidate inhibitors of ZIKV replication. The aim of this work was to investigate ZIKV propagation and quantification in different cell lines. The human (U87, A549, Huh7), mosquito (C6/36) and monkey (VERO E6) cell lines tested were all permissive to ZIKV infection. When assessed by plaque forming units (PFU) in three different target cell lines, the maximal production of ZIKV was achieved in Huh7 at day 3 post-infection (6.38 ± 0.44 log10 PFU/ml). The C6/36 cell line showed a low and slow production of virus when compared with other cell lines. A549 readout cells generated a larger number of plaques compared to Huh7 but not to VERO E6 cells. ZIKV PFU and RNA titers showed the highest correlation when Huh7 and A549 were used as the producer and readout cells, respectively. Also, U87 cells produced ZIKV RNA titers which were highly correlated with PFU independently from the readout cell line. Using the best virus-cell system, sofosbuvir and ribavirin EC50 were 1.2 μM and 1.1 μM when measured through plaque assay, and 4.2 μM and 5.2 μM when measured by quantitative real time PCR (qRT-PCR), respectively. In summary, ZIKV can efficiently infect different human cell lines and rapidly reach peak viral titers. Overall, A549 cells appear to be as efficient as the VERO E6 gold standard for plaque assay allowing the use of human, rather than simian, cells for evaluating candidate anti-ZIKV compounds by the reference assay. The possibility to replace the labor-intensive plaque assay with the more rapid and easy-to-perform qRT-PCR is appealing and warrants further investigation.

Introduction

Zika virus (ZIKV) is a member of the Flaviviridae family, which includes several agents of clinical significance, such as Dengue, Hepatitis C (HCV), West Nile and Japanese encephalitis viruses. ZIKV is assumed to be propagated in nature through an enzootic cycle involving mammalian hosts, such as primates, and insect vectors, primarily mosquitoes of the Aedes genus (Akoua-Koffi et al., 2001, Fagbami, 1979, Marchette et al., 1969, McCrae and Kirya, 1982). ZIKV can be transmitted to humans by the bite of female Aedes mosquitoes, through a sexual route, from a pregnant mother to her unborn fetus, by blood transfusion, or other bodily fluids (Barzon et al., 2016, Miner et al., 2016, Petersen et al., 2016, Sharma and Lal, 2017). Three geographically distinct lineages of ZIKV have been reported, including the East African, West African, and Asian (Baronti et al., 2014). This last one seems to have evolved from the African lineage and has been shown to have increased pathogenicity and epidemic potential (Lazear and Diamond, 2016, Wang et al., 2016, Weaver et al., 2016). Up to 80% of human ZIKV infections appear to be asymptomatic, with a small subset of cases presenting with mild flu-like symptoms associated with rash, arthralgia, and conjunctivitis (Lessler et al., 2016, Marano et al., 2016, Shah and Kumar, 2016). However, the recent spread across the Brazilian territory and the neighboring countries in 2015 has renewed the interest in ZIKV due to the association between infection acquired during pregnancy and congenital malformations, including microcephaly (Brasil et al., 2016a, de Araújo et al., 2016, Paploski et al., 2016, Rasmussen et al., 2016), spontaneous abortion and intrauterine growth restriction (Carteaux et al., 2016, Miner et al., 2016). In addition, a broad range of neurological disorders have been reported in adults, including Guillain-Barré syndrome (GBS) (Brasil et al., 2016b, Cao-Lormeau et al., 2016, Oehler et al., 2014). Phylogenetic analysis revealed that the ZIKV strains currently circulating in South America share >99% identity with ZIKV isolates from French Polynesia outbreaks (Asian Lineage) (Imperato, 2016) and seem to be associated with neurologic damage, including few cases of bilateral macular and perimacular lesions (Mlakar et al., 2016, Ventura et al., 2016), globally proving that ZIKV genetic evolution has resulted in expanded virus tropism and increased pathogenicity. The emergence of ZIKV associated pathology in Brazil led the WHO to declare the ZIKV outbreak to be a public health emergency of international concern (WHO, 2016).

Despite the clinically relevant consequences of ZIKV in humans, currently there are no countermeasures to halt transmission, control infection or mitigate disease (Bollati et al., 2010, Yun and Lee, 2017). Some broad-spectrum antivirals, such as interferons (IFNs), ribavirin and favipiravir, cannot be used against ZIKV because they can be harmful to pregnant women (Barrows et al., 2016, Chutaputti, 2000, Xu et al., 2016). Recently identified crosstalk pathways between cancer and viral replication have drawn the attention on FDA-approved cancer drugs which show anti-ZIKV activity (Cheng et al., 2016). The repurposing of these “old” drugs to identify compounds with novel activity against ZIKV infection can be a useful tool to overcome the high cost of antiviral drug-discovery pipeline. In addition, the high degree of conservation of viral RNA polymerase among the Flaviviridae family has recently prompted the evaluation of compounds approved for treatment of HCV infection. Indeed, the nucleotide analog sofosbuvir, approved in 2013, has shown anti-ZIKV activity in vitro, although with effective concentration significantly higher than those reported with HCV (Bullard-Feibelman et al., 2017, Murakami et al., 2010, Sacramento et al., 2017). To evaluate candidate anti-ZIKV compounds, robust methods for ZIKV propagation and quantification are required. Monkey (VERO E6) or insect (C6/36) cell lines have been most frequently used to propagate ZIKV and test candidate antivirals (Adcock et al., 2017, Goebel et al., 2016, Pascoalino et al., 2016). Interestingly, cell-dependent inhibition of ZIKV replication has been shown for sofosbuvir (Eyer et al., 2016, Mumtaz et al., 2017, Sacramento et al., 2017). This advises for investigating multiple ZIKV propagation protocols to represent the variability of human cells to process candidate anti-ZIKV compounds and impact their activity.

In this work, we have evaluated the kinetics of viral replication in several cell lines, as measured by both ZIKV RNA and replication competent virus through plaque assay, to define the best standards for ZIKV antiviral screening. As a proof of concept, the inhibitory effect of sofosbuvir and ribavirin on ZIKV has been assessed through a yield reduction assay.

Section snippets

Cells

ZIKV growth was evaluated in VERO E6 (African green monkey kidney; ATCC® CRL-1586™), C6/36 (Aedes albopictus mosquito; ATCC catalog no. CRL-1660) and in three different human cell lines: A549, derived from lung carcinoma (ATCC® CCL-185™), Huh7, derived from human hepatoma (kindly provided from Istituto Toscano Tumori, Core Research Laboratory, Siena, Italy) and U87, a glioblastoma cell line (NIBSC 0044). The cell propagation medium was Dulbecco's Modified Eagle's Medium High Glucose with sodium

ZIKV propagation in different cell lines

All the cell lines evaluated were permissive to ZIKV infection. Depending on cell type the peak of infectivity was reached at different time points, as measured by plaque assay in A549, Huh7 and VERO E6 cells (Fig. 1). On average, the maximal production of infectious ZIKV was reached in Huh7 at day 3 (6.38 ± 0.44 log PFU/ml). In the A549 cell line, the peak of infectivity was reached at day 2 (5.85 ± 0.19 PFU/ml), significantly before VERO E6 and U87 (5.51 ± 0.46 and 5.91 ± 0.23 log10 PFU/ml at day 4,

Discussion

Since the Brazilian ZIKV outbreak, many efforts have been directed to find new or old drugs able to prevent the serious consequences of ZIKV infections and the mother-to-child transmission. In vitro screening and analysis of the mechanism of action of candidate antiviral drugs is best performed by using different cell types, particularly human targets, which are more representative for in vivo drug therapy.

Very few methodological studies have been conducted to assess the best conditions of ZIKV

Acknowledgements

This work was partially supported by the Project UNAVIR “A novel strategy to combat multiple viral diseases with one antiviral agent” (Bando FAS Salute 2014 – Regione Toscana). The funding sources had no involvement in study design; in the collection, analysis and interpretation of data; in the writing of the report; and in the decision to submit the article for publication.

We thank Dr. Giulietta Venturi (Istituto Superiore della Sanità), for gently provided the ZIKA virus strain used in this

References (48)

  • A.W.R. McCrae et al.

    Yellow fever and Zika virus epizootics and enzootics in Uganda

    Trans. R. Soc. Trop. Med. Hyg.

    (1982)
  • J.J. Miner et al.

    Zika virus infection during pregnancy in mice causes placental damage and fetal demise

    Cell

    (2016)
  • N. Mumtaz et al.

    Cell-line dependent antiviral activity of sofosbuvir against Zika virus

    Antiviral Res.

    (2017)
  • E. Murakami et al.

    Mechanism of activation of PSI-7851 and its diastereoisomer PSI-7977

    J. Biol. Chem.

    (2010)
  • C.V. Ventura et al.

    Zika virus in Brazil and macular atrophy in a child with microcephaly

    Lancet

    (2016)
  • L. Wang et al.

    From mosquitos to humans: genetic evolution of zika virus

    Cell Host Microbe.

    (2016)
  • S.C. Weaver et al.

    Zika virus: history, emergence, biology, and prospects for control

    Antiviral Res.

    (2016)
  • T.V.B. de Araújo et al.

    Association between Zika virus infection and microcephaly in Brazil, January to May, 2016: preliminary report of a case-control study

    Lancet Infect. Dis.

    (2016)
  • D.S. Agbulos et al.

    Zika virus: quantification, propagation, detection, and storage

    Curr. Protocols Microbiol.

    (2016)
  • C. Akoua-Koffi et al.

    Investigation surrounding a fatal case of yellow fever in Côte d’Ivoire in 1999

    Bull. Soc. Pathol. Exot.

    (2001)
  • C. Baronti et al.

    Complete coding sequence of zika virus from a French Polynesia outbreak in 2013

    Genome Announc.

    (2014)
  • L. Barzon et al.

    Isolation of infectious Zika virus from saliva and prolonged viral RNA shedding in a traveller returning from the Dominican Republic to Italy, January 2016

    Eur. Surveill. Dis. Bull.

    (2016)
  • P. Brasil et al.

    Zika virus infection in pregnant women in rio de janeiro – preliminary report

    N. Engl. J. Med. N. Engl. J. Med.

    (2016)
  • G. Carteaux et al.

    Zika virus associated with Meningoencephalitis

    N. Engl. J. Med.

    (2016)
  • Cited by (42)

    • System-oriented optimization of multi-target 2,6-diaminopurine derivatives: Easily accessible broad-spectrum antivirals active against flaviviruses, influenza virus and SARS-CoV-2

      2021, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Cells were incubated at 37 °C in a humidified incubator supplemented with 5% CO2. All the viral stocks were titrated by plaque reduction assay (PRA) as previously described [65]. Briefly, confluent Vero E6 (WNV, DENV, SARS-CoV-2) and A549 (ZIKV) in 6-well format were infected with three tenfold dilutions of viral stock and after 1 h adsorption at 37 °C, the cells were washed with PBS and 0.75% Sea Plaque Agarose (Lonza) was added to each well.

    • Labyrinthopeptin A1 inhibits dengue and Zika virus infection by interfering with the viral phospholipid membrane

      2021, Virology
      Citation Excerpt :

      We selected Vero and A549 cell lines because they express the phosphatidylserine receptors TIM-1 and AXL, and chose Raji DC-SIGN cells because they express the DC-SIGN receptor. All these receptors are involved in DENV as well as ZIKV infection, and Flavivirus infection in these cell lines has been extensively studied by our and several other research groups (Alen et al., 2009; Himmelsbach and Hildt, 2018; Vicenti et al., 2018). Some additional cell lines that are biologically more relevant for either DENV or ZIKV infection were included as well.

    • Inhibition of Brazilian ZIKV strain replication in primary human placental chorionic cells and cervical cells treated with nitazoxanide

      2020, Brazilian Journal of Infectious Diseases
      Citation Excerpt :

      Corroborating our data, Thaker et al. demonstrated that ZIKV infection differentially reprograms the host cell glucose metabolism, leading to cell death in human but not in C6/36 mosquito cells.33 Regarding infection rate, studies have reported that ZIKV BR isolates display differential abilities to infect lineages or primary mammalian cells and insect cells.34,35 Moreover, mammalian cells also showed different susceptibility rates when infected with the Asian and African ZIKV lineages, and infection with ZIKV prototypes (MR766 and PE243) induces greater protein expression and viral progeny than ZIKV BR isolates.34

    View all citing articles on Scopus
    View full text