INTRODUCTION
Recent outbreaks with (re)emerging viruses such as Zika virus (ZIKV), dengue virus (DENV), Ebola virus, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) illustrate the enormous threat viruses pose to our global health system. ZIKV and DENV, among other pathogenic viruses such as yellow fever virus and West Nile virus, are positive-sense single-stranded RNA viruses belonging to the flaviviruses (genus:
Flavivirus). ZIKV has been circulating for many decades without causing major threats but transformed to a “Public Health Emergency of International Concern” in 2015, when it caused a severe epidemic in South America (
1). Its association with fetal microcephaly, as well as with other neurologic complications in adults, makes it a serious threat (
2–4). Novel outbreaks of ZIKV remain possible since its transmission vector,
Aedes mosquitoes, is able to increasingly spread to new areas due to several factors such as global warming and urbanization (
5). Despite many promising efforts, no antiviral drug or vaccine to treat or prevent ZIKV infection has made it to the market yet.
Traditional methods to identify potential antiviral compounds are often based on the evaluation of cytopathic effects (CPE) in host cells after viral infection. These include the plaque assay, 50% cell culture infectious dose (CCID
50) determination, or the determination of cell viability using colorimetric detection with, for example, the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay (
6–8). The potency of the compounds can also be determined by assessing viral replication with quantitative PCR or by detecting viral antigens using immunofluorescence (
9,
10). Assays using recombinant viruses that express reporter proteins are also commonly used, but to this end, viruses have to be genetically modified (
11,
12). Usually, these assays are performed as endpoints that are labor intensive and/or that require complex analysis. Label-free real-time technologies are therefore attractive alternatives in the search for potential antiviral compounds.
Cell-based electrical impedance (CEI) is a label-free real-time technology that measures changes in impedance (
Z) of a cell layer grown on a surface with embedded (gold) electrodes. The cells are exposed to an electric field generated by continuous sweeping of noninvasive alternating current (AC) voltages over a range of frequencies. As cells act as insulating particles in this system, they will resist the flow of AC, resulting in a frequency-dependent resistance measurement, also called impedance (
Z). Changes in cell morphology, growth, or adhesion due to altered cell viability, migration, growth, spreading, proliferation, or any other change can lead to the current being more or less impeded. CEI has gained popularity in recent years and is used to monitor dynamic responses of cells toward receptor ligands, drugs, and pathogens. In this manner, CEI has been successfully implemented in cytotoxicity studies, cancer cell behavior, signaling pathway elucidation, and endothelial barrier function (
13,
14).
Since many viruses cause CPE in susceptible cells, CEI can also be used in antiviral research to complement conventional antiviral assays. As cells grow, they spread out over the instrument’s electrodes. The growth of cells on the electrodes, together with the formation of tight junctions, will impede the current flow and subsequently will increase the measured impedance. On the other hand, as cells start to deform or detach due to viral infection, tight junctions will be disrupted, and the electrode surface will be covered with fewer cells or more loosely attached cells. This will allow more current passage and in turn will decrease the measured impedance (
15–17). As such, CEI has been used to replace traditional CCID
50 or plaque assays in virus titer determination (
18). CEI has been implemented previously in antiviral research with various human and animal viruses such as influenza A virus, several herpesviruses, chikungunya virus, and SARS-CoV-2 (
19–24). It can measure virus-induced changes and the activity of antivirals or neutralizing antibodies in real time, in a label-free manner, and in a medium-throughput setting. Nevertheless, the technology has been underexploited in the search for
Flavivirus inhibitors. Fang et al. used CEI to study CPE induced by West Nile virus and St. Louis encephalitis virus and to quantify the protecting role of neutralizing antibodies (
17). Cheng et al. determined that DENV-induced CPE was observed earlier with CEI than with conventional microscopy (
25). However, to our best knowledge, the consequences of ZIKV infection on cellular impedance and the evaluation of ZIKV inhibitors have never been studied using CEI.
In the present study, we use electric cell-substrate impedance sensing (ECIS), an established CEI technology originally developed by Giaever and Keese (
26,
27), to monitor cell growth and antiviral activity of various compounds after ZIKV (or DENV) infection in real time. We use ECIS to evaluate several compounds with described anti-ZIKV activity: the entry inhibitors labyrinthopeptin A1 (Laby A1) and duramycin (
28,
29), as well as the polymerase inhibitors NITD008 (
30) and 7-deaza-2′-
C-methyladenosine (7DMA) (
31). We compare the obtained ECIS results with traditional antiviral assays based on cell viability or virus yield to validate CEI as an antiviral tool. Furthermore, we use ECIS to determine various parameters to characterize the
in vitro activity profile of antiviral compounds in more detail. Hereby, we validate CEI as a powerful tool to monitor ZIKV infection and to decipher the antiviral activity of compounds.
DISCUSSION
In this study, we demonstrate the power of implementing cell-based electrical impedance measurements in the search for Flavivirus inhibitors. The CEI assay offers valuable information when monitoring morphological cell changes in response to ZIKV infection in real time, in a label-free manner, and in a noninvasive assay format. Impedance measurements can be used to quantify viral load, and the kinetic readout can help determine at which time points endpoint assays should be performed. We used CEI to—for the first time—determine the antiviral activity of a set of previously described ZIKV entry and replication inhibitors. The assay can be used to determine compound cytotoxicity, as well as the compound’s dynamic antiviral activity profile, and this with the same metric. Finally, we also aimed to categorize compounds based on impedance features that might be representative of their mechanism of action. Additional sets of compounds should be tested in the future to assess whether the assay can indeed be used to differentiate between entry blockers and later stage antivirals.
Phenotypic assays are routinely used in antiviral research by both academic groups and the pharma industry. In general, the reduction in CPE or plaque formation after compound treatment is evaluated, for example, microscopically or with a luminescent, fluorescent, or colorimetric readout. However, these endpoint assays are very often time consuming and possess other drawbacks; they require multiple handling steps, the onset of CPE or the kinetics of the infection cannot be detected, and it is crucial that the optimal endpoint is chosen in order to achieve a good assay quality. CEI, on the contrary, offers a simple noninvasive and label-free workflow, with real-time measurements and an objective/unbiased readout. Furthermore, the samples can be further processed after the impedance experiment is finished; since the method is noninvasive, qPCR can be performed on both cells and supernatant fractions, while the cells can also be lysed for Western blot (WB) analysis. Moreover, after the impedance run, staining can be performed, and the plates can be imaged. The usefulness of the CEI technology in the search for antivirals has been reported previously for other pathogenic viruses (
19,
20,
22,
23). The main limitation of the ECIS assay (and other CEI assays) is related to the equipment needed: the ECIS device consists of a well station that is placed in a cell culture incubator, a control module, and a computer equipped with the analyzing software. Furthermore, specific microtiter plates embedded with gold electrode sensors are required, increasing the cost of the assay. The instrument’s throughput might also represent an issue. During the whole duration of the experiment, no other assays can be run. Of course, the experiment can also be run as an endpoint assay to increase throughput, by measuring the samples’ impedance only during the time window of interest. Hence, kinetic data are not provided. Because of the rather low throughput, CEI is not attractive in a compound screening setting. Including sufficient controls and replicates, up to 45 compound samples can be monitored simultaneously on a 96-well format. However, since CEI is becoming increasingly popular, the ECIS equipment’s throughput might by increased in the future. Agilent, another provider of CEI technology (xCELLigence RTCA), already provides 384-well formats. Despite the throughput, CEI has other valuable applications in a preclinical research. We suggest that it could be implemented either in an initial phase to facilitate high-throughput screening (e.g., by determining interesting time points) and in later stages to complement current methodologies during further selection and characterization of novel antivirals (i.e., during lead optimization and mechanism of action studies).
More recent models of the ECIS instrument allow the direct measurement of the two impedance components: resistance and capacitance. These are interesting parameters as they provide additional biological information about the different cellular behaviors at play, with resistance being more prone to changes in cell-cell contacts, while capacitance rather represents cell-substrate interactions and cell surface coverage (
16). When both parameters are monitored, this could potentially reveal additional information on ZIKV infection biology, in addition to the currently described CPE measurement.
Impedance-based biosensors are also currently emerging to complement traditional diagnostic tools in virology such as enzyme-linked immunosorbent assay (ELISA) and RT-qPCR (
43,
44). Here, impedance measurements also have some advantages over the more traditional assays, as they provide a sensitive, label-free, and real-time readout without requiring laborious sample preparation. For diagnostic purposes, a target-specific biorecognition element is often implemented in the sensor. Impedance-based biosensors have been developed for the specific detection of ZIKV or other flaviviruses, where virus-specific capture antibodies or probes have been immobilized to the electrodes of the sensor (
45–47). However, the course of
in vitro ZIKV infection and the evaluation of inhibitors have never been studied. Therefore, this study further demonstrates the capabilities of this technology for virus studies, while also adding new insights to
Flavivirus antiviral research.
Using the CEI assay, we identified PRO2000 as a novel ZIKV inhibitor. The assay also indicated that PRO2000 has pronounced effects on the cell monolayer, albeit in a nontoxic way. Previous research has shown that PRO2000 potentially has multiple mechanisms of action against HIV, since it—in addition to interacting with HIV glycoprotein gp120—also interacts with the CD4 and CXCR4 cell membrane receptors, both HIV entry receptors (
48). It has also been shown previously that PRO2000 inhibits cell-to-cell transmission of HTLV-1 virus (
49). PRO2000 is a polyanionic polymer, indicating that the negative charges could possibly interact with positive charges of various cell surface proteins. These cellular interactions and multiple targets could explain the altered impedance response that we observed. Although our results show that PRO2000 has pronounced effects on the cells’ behavior
in vitro, clinical trials with PRO2000 have demonstrated that it is safe for therapeutic use in humans (
50).
Like previous studies that have used CEI for virus research (
16,
19,
23), we further demonstrate how this label-free kinetic readout can complement more traditional methods and more specifically how it can help researchers to better evaluate their selected lead agents. Our work demonstrates that when different methods complement each other, new insights in the field of antiviral research can be gained.
MATERIALS AND METHODS
Cell lines, primary cells, and virus strains.
(i) Cell lines. Human lung carcinoma A549 cells and baby hamster kidney (BHK-21) cells were obtained from ATCC (Manassas, VA, USA) and grown in minimum essential medium (MEM; Thermo Fisher Scientific [TFS], Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS), 2 mM l-glutamine (TFS), and 0.075% sodium-bicarbonate (TFS). The mosquito cell line C6/36 (isolated from Aedes albopictus) was obtained from ATCC and cultured in Leibovitz’s L-15 medium (TFS) supplemented with 10% FBS, 0.01 M HEPES (TFS), and penicillin/streptomycin (TFS). Cell cultures were maintained at 37°C in a humidified environment with 5% CO2, except for C6/36 cells, which were cultured at 28°C in the absence of CO2. The cells were passaged every 3 to 4 days.
(ii) Viruses. ZIKV prototype strain MR766 (isolated from sentinel Rhesus monkey, Uganda, 1947) was obtained from ATCC. ZIKV was propagated in C6/36 cell cultures from which supernatant containing the virus was harvested 5 to 9 days after infection and stored at −80°C. Viral titers were determined using plaque assays in BHK-21 cells, as described below. All viruses were obtained and used as approved according to the rules of a Belgian institutional review board (Departement Leefmilieu, Natuur en Energie, protocol SBB 219 2011/0011n) and the Biosafety Committee at the Katholieke Universiteit Leuven.
Antiviral test compounds.
Labyrinthopeptin A1 (2,073.7 Da) was isolated and purified as described earlier (
51). Duramycin and NITD008 were purchased from Sigma-Aldrich (St. Louis, MO, USA). 7-Deaza-2′-C-methyl-
d-adenosine (7DMA) was purchased from Carbosynth (Berkshire, UK). PRO2000 (molecular weight: ~5,000 g/mol) was kindly provided by A.T. Profy (formerly at Indevus Pharmaceuticals Inc., Lexington, MA, USA).
CPE reduction and cytotoxicity assays.
Test compounds were screened for their antiviral activity using a colorimetric antiviral assay that was originally described by Pauwels et al. (
52). We adapted the protocol as described in more detail by Van Hout et al. (
53). A549 cells were seeded in cell culture medium in a 96-well plate at 15 × 10
3 cells/well and allowed to adhere and grow overnight. The next day, serial dilutions of compound were prepared in cell culture medium without FBS (final FBS concentration was 4%), followed by 30 min of incubation. Then, ZIKV was added at a multiplicity of infection (MOI) of 1. Negative controls (cell controls [CCs]) were mock infected with culture medium and DMSO. After 3 incubation days, supernatant was collected and stored at −80°C for further analysis with RT-qPCR. CPE was determined microscopically, and the cell viability was determined using the spectrophotometric MTS/phenazine ethosulfate (PES) viability staining assay (Cell-Titer 96 Aqueous one solution Proliferation assay kit; Promega, Madison, WI, USA). Absorbance was measured at 498 nm using the Versamax microplate reader and analyzed using SoftMax Pro software (Molecular Devices, Sunnyvale, CA, USA). To determine whether the compound itself induced cellular toxicity, the assays were also performed without the addition of virus. The 50% inhibitory concentration (IC
50), which is defined as the compound concentration that is required to inhibit virus-induced CPE by 50%, and the 50% cytotoxic concentration (CC
50), which is defined as the compound concentration required to reduce the cell viability by 50%, were determined. Each experiment was performed in duplicate.
ECIS assay.
The electric cell-substrate impedance sensing (ECIS) Z array station (Applied Biophysics, Troy, NY, USA) was used to measure changes in electrical resistance across the cell monolayer during incubation with compounds and viral infection. ECIS plates with interdigitated electrodes (96W20idf PET) were washed prior to use with culture medium for 4 h at 37°C. Next, medium was replaced by an A549 cell suspension (15 × 10
3 cells/well). The growth of the cells was monitored overnight in the ECIS Z array station in multifrequency (MFT) mode at 37°C in 5% CO
2. In MFT mode, the electrodes are probed with a weak and noninvasive alternating current (AC) signal at 11 frequencies between 10 and 10
5 Hz to measure the frequency-dependent impedance (
Z). Changes in
Z that are representative for cell adhesion and proliferation were measured for 24 h. The next day, compound dilutions were added in culture medium without FBS (final FBS concentration was 4%), followed by infection with ZIKV MR766 at various MOI. Negative controls (CC) were mock infected with culture medium and DMSO. The measurement was continued in MFT mode for the subsequent 5 days. Every ECIS experiment lasted 160 h in total: 24 h with cells only and the remaining time with compound and virus. Measurements were taken every 8 min. The data were further processed using Microsoft Excel (Microsoft, Redmond, WA, USA). During every experimental run, each sample was measured through time and in duplicate, and the mean
Z ± SD at every time point was calculated. Impedance is reported at 16,000 Hz, because at this frequency, the difference in impedance between VC and CC is most pronounced 3 days postinfection (
Fig. 1A).
Viral plaque assay.
BHK-21 cells were seeded in 12-well plates in growth medium (4 × 105 cells/well). The next day, the cells were incubated in triplicate with either 10-fold viral stock dilutions or with cell supernatant dilutions collected at different hours postinfection. Assay medium was included as a negative control. After 1 h, medium was replaced with a microcrystalline cellulose overlay (Avicel RC 581, IMCD Benelux, Mechelen, Belgium) and incubated for 4 days. The overlay was removed, after which the cells were fixed in 70% ethanol and stained with crystal violet solution (Merck, Darmstadt, Germany). The plaques were counted, and the infectious viral titer was determined according to the following formula: number of plaques × dilution factor × (1/inoculation volume).
RNA isolation and quantitative RT-PCR.
Virus lysis, RNA isolation, and RT-qPCR were performed using the CellsDirect one-step RT-qPCR kit (Thermo Fisher Scientific) according to the manufacturer’s instructions. During RT-qPCR, the ZIKV E protein encoding region (nucleotides 1193 to 1269) was amplified using the primers 5′-
CCGCTGCCCAACACAAG-3′ (forward) and 5′-
CCACTAACGTTCTTTTGCAGACAT-3′ (reverse), together with a Double-Quenched Probe 5′-6-FAM/
AGCCTACCT/ZEN/
TGACAAGCAATCAGACACTCAA/3′ IBFQ (
54,
55). Primers and probes were obtained from Integrated DNA Technologies (IDT, Leuven, Belgium). The viral copy numbers were quantified based on a standard curve produced using serial 10-fold dilutions from viral DNA templates with known concentrations.
Real-time IncuCyte assay.
A549 cells were seeded (15 × 103 cells/well), and after overnight incubation, they were infected with ZIKV MR766 at various MOIs or left untreated. The cells were incubated and imaged in real time at 37°C for 4 days in an IncuCyte S3 (Essen BioScience Inc., Ann Arbor, MI, USA). Images were taken every 8 h, with five fields imaged per well under ×4 magnification.
Data analysis.
The impedance data were normalized (
Z′) according to the following formula:
The impedance at the time just prior to infection (
Z0 hpi) was set to a value of 1, and the impedance at the end of the experiment (160 h;
Zend) was set to a value of 0, while the impedance of the intervening time points (
Zx) were scaled relative to
Z0 hpi and
Zend. GraphPad Prism (version 9.2.0, GraphPad Software Inc., San Diego, CA, USA) was used to fit the normalized data into a curve. When quantifying the compounds’ antiviral activity against ZIKV, various parameters were calculated to allow evaluation and comparison of the compound potencies: CIT
50, AUC
n, and IC
50. CIT
50 was calculated as the time point at which impedance had decreased 50%, compared to the maximum impedance of the cell control. The area under the normalized curve (AUC
n) was calculated over the whole duration of the experiment (160 h). This parameter represents the growth and viability of the cells after seeding, treatment, and/or infection. The percentage inhibition of a compound at a particular concentration in both ECIS and CPE reduction assays was obtained by subtracting the negative-control (cell control [CC]) response followed by normalizing to the positive-control (virus control [VC]) response. Dose-response curves were obtained with GraphPad Prism using the nonlinear regression four-parameter fitting tool. IC
50 values were used to represent the potency of the compounds and calculated according to the following formula:
Here, C1 is the compound concentration resulting in more than 50% inhibition, C2 is the compound concentration resulting in less than 50% inhibition, with their respective percentage of inhibition (%inhib
C1/C2). To compare IC
50 values or Hill slopes, two-tailed unpaired
t tests were used.
P values < 0.05 were considered significant. The quality of the ECIS assay was determined using the
Z′ factor, based on AUC
n values between 0 and 160 h after seeding (
19,
56). This was calculated using following formula:
where σ
vc and σ
cc correspond to AUC
n standard deviations of VC and CC, respectively; and μ
vc and μ
cc correspond to AUC
n means of VC and CC, respectively. All performed assays in our study had
Z′ factors above 0.5, which is considered an excellent screening assay value (
56) (data not shown).
ACKNOWLEDGMENTS
We thank M. Jacquemyn for excellent guidance when using the IncuCyte.
This work was supported by KULeuven grant PF/10/018, by Fonds voor Wetenschappelijk Onderzoek grant G.485.08, and by the Fondation Dormeur Vaduz.
We declare no conflict of interest.
M.O.: conceptualization, methodology, investigation, visualization, writing; E.M.: investigation; J.D.: conceptualization, methodology, writing; D.S.: supervision, writing.