Skip to main content
  • Original article
  • Open access
  • Published:

Establishment of in-house assay for screening of anti-SARS-CoV-2 protein inhibitors

Abstract

Developing a potent antiviral agent to combat Coronavirus Disease-19 (COVID-19) is of critical importance as we may be at risk of the emergence of new virus strains or another pandemic recurrence. The interaction between the SARS-CoV-2 spike protein and Angiotensin Converting Enzyme 2 (ACE2) is the main protein-protein interaction (PPI) implicated in the virus entry into the host cells. Spike-ACE2 PPI represents a major target for drug intervention. We have repurposed a previously described protein-protein interaction detection method to be utilized as a drug screening assay. The assay was standardized using Chitosan nanoparticles (CNPs) as the drug and SARS-CoV-2 spike-ACE2 interaction as the PPI model. The assay was then used to screen four natural bioactive compounds: Curcumin (Cur), Gallic acid (GA), Quercetin (Q), and Silymarin (Sil), and their cytotoxicity was evaluated in vitro. Production of the spike protein and the evaluation of its activity in comparison to a standard commercial protein was part of our work as well. Here we describe a novel simple immunofluorescent screening assay to identify potential SARS-CoV-2 inhibitors that could assess the inhibitory effect of any ligand against any PPI.

Graphical Abstract

Introduction

Coronavirus,the virus that caused the 2020 pandemic, also known as severe acute respiratory syndrome Coronavirus-2 (SARS-CoV-2) has significantly phased out. However, the efforts to develop and repurpose drugs to be used for the treatment of Coronavirus disease-19 (COVID-19) are still in progress (Lai and Hsueh 2023; Murakami et al. 2023; Saravolatz et al. 2023). To date, the choices for antivirals against SARS-CoV-2 are very limited with only a few FDA-approved drugs. Remdesivir is a broad-spectrum antiviral that inhibits viral replication by terminating ribonucleic acid (RNA) transcription. It was the first drug to be approved in 2020 for the treatment of hospitalized or non-hospitalized patients at high risk for COVID-19 disease progression (Lamb 2020). Following Remdesivir, Tocilizumab (Actemra®) and Baricitinib (Olumiant®), immunosuppressive drugs used for the treatment of rheumatoid arthritis, were approved in 2022 for the treatment of COVID-19 in hospitalized adults who are receiving systemic corticosteroids and require supplemental oxygen, ventilation, or extracorporeal membrane oxygenation (ECMO) (Assadiasl et al. 2021; Bozorgmanesh et al. 2021).

Spike protein is a surface protein of the SARS-CoV-2 virus that mediates viral adhesion and fusion by interaction with the angiotensin-converting enzyme 2 (ACE2) receptor expressed on the surface of the host cells (Scialo et al. 2020). Spike-ACE2 protein-protein interaction (PPI) is a significant step in virus replication and has been proposed as a target for the discovery of antiviral agents (Jia et al. 2021).

Many research groups have sought to inhibit this PPI using different natural compounds and drugs. Screening methods adopted by several research teams varied, but many used commercial ELISA kits, a sensitive and relatively affordable method (Ahamad et al. 2022; Bojadzic et al. 2021; Tedesco et al. 2022). However, importing such kits to low-income countries is unaffordable and time-consuming.

For this reason, we focused on developing an in-house immunofluorescent assay to screen several inhibitors, with the possibility of repurposing this assay for the screening of ligands, e.g., small molecules, against any other PPI. We sought to exploit a protein-protein interaction detection method that we have developed to detect spike-ACE2 PPI which allowed us to evaluate the binding kinetics of that PPI (Emam et al. 2022). To further increase the affordability of our assay, we opted to produce the spike protein in the lab and use it in the screening assay.

Natural bioactive compounds such as phytochemicals are usually preferred as a treatment option, when applicable, due to their relatively few side effects and sustainable production. Many efforts were made to investigate herbs, herbal extracts, or active compounds for anti-COVID-19 activity (Al-Harrasi et al. 2022; Bizzoca et al. 2022; Chakravarti et al. 2021; Khan et al. 2021). In a previous report, we demonstrated that silymarin, which is a flavonolignan found in Silybum marianum (Milk thistle), has an antiviral effect against SARS-CoV-2 and speculated that its mechanism of action may be blocking the viral entry (Loutfy et al. 2022). Gallic acid is a bioactive compound classified as phenolic acid found in many plant families. Gallic acid has shown anti-hepatitis C virus (HCV) and anti-herpes simplex virus-2 (HSV-2) activities in previous literature (Govea-Salas et al. 2016; Jadel Müller Kratz et al. 2008). Quercetin (Q) is a flavonol, which is a polyphenol compound found in plants, with a wide pharmacological activity spectrum ranging from anti-viral and anti-carcinogenic activity to general health improvement and psycho-stimulation ascribed to its potent antioxidant effect (Li et al. 2016; Xu et al. 2019). Quercetin showed an ability to bind to spike protein and ACE2 in silico (Hiremath et al. 2021; Pan et al. 2020). Curcumin (Cur) is an active compound derived from Curcuma longa. In several clinical trials, it was found to ameliorate inflammation in COVID-19 patients, and it was also proposed to be used as an adjuvant drug in COVID-19 treatment (Rattis, Ramos, and Celes 2021; Vahedian-Azimi et al. 2022).

Here, we have extended the application of our previously published protein-protein interaction detection method (Emam et al. 2022), to screen these selected compounds. The assay was standardized using chitosan nanoparticles (CNPs), a known anti-COVID-19 compound (Loutfy et al. 2022), and the spike-ACE2 interaction model. It was applied to screen the four promising natural compounds: Curcumin (Cur), Gallic acid (GA), Quercetin (Q), and Silymarin (Sil), to elucidate their potential inhibitory activity against spike-ACE2 PPI. We also produced the spike protein whose activity was evaluated and compared with the commercial protein.

Materials and methods

Cell lines

Human Embryonic Kidney cells (HEK293T) (ATCC® CRL-3216™) cells and African green monkey kidney (Vero) (ATCC® CCL-81 ™) cells were maintained in Dulbecco’s modified Eagle’s medium (DMEM) (Thermo Fisher Scientific, Gibco™) with 10% FBS (Thermo Fisher Scientific, Gibco™) and 2% Penicillin/Streptomycin (Invitrogen, Inc., Carlsbad, CA).

Reagents and antibodies

Curcumin (Cur), Gallic acid (GA), Quercetin (Q), Silymarin (Sil), (3-aminopropyl) triethoxysilane (APTES, ≥ 98%), EDC N-(3-dimethylaminopropyl)-N0-ethylcarbodiimide hydrochloride, and N-hydroxy succinimide (NHS) were all purchased from Sigma-Aldrich, USA. MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) dye and dimethyl sulfoxide (DMSO) were purchased from Serva Electrophoresis GmbH, Germany. Lipofectamine 3000 reagent (Thermo Fisher Scientific, Invitrogen™) was used for transfection. Spike protein ectodomain encoding plasmid, a generous gift from Dr. Jason S. McLellan (Wrapp et al. 2019), was purified with PureLink™ HiPure Plasmid Maxiprep Kit (K210006, Thermo Fisher Scientific, Invitrogen™). The antibodies used were as follows: Rabbit anti-spike antibody (40,591-T62, Sino biological), spike monoclonal antibody (E-AB-V1008, Elabscience), Goat anti-rabbit antibody (ab6721, Abcam), and Goat anti-human IgG FITC (E-AB-1019, Elabscience). Protein production and quantification were performed by Strep-Tactin®XT Spin Column (2-4151-000, iba-lifesciences) and BCA assay kit (23,225, Pierce). Recombinant human ACE2 (PKSH032068, Elabscience) and Recombinant 2019-nCoV spike (PKSR030477, Elabscience) were used in drug screening.

Plasmid preparation and transfection

The plasmid expressing C-terminal Twin strip tagged spike protein was amplified and purified with a maxiprep kit following the manufacturer’s instructions. HEK293T cells were seeded in a 6-well plate, 6 × 105 cells per well, in DMEM complete media. The following day, the cells were transfected with the plasmid using Lipofectamine 3000 reagent, following the manufacturer protocol. The cells were harvested 72 h post-transfection and prepared for sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis.

Western blot

For the preparation of cell lysates, cells were washed twice with ice-cold PBS, harvested, re-suspended in Laemmli’s buffer, and boiled for 5 min. Samples were separated on 9% SDS‐PAGE and transferred onto polyvinylidene difluoride (PVDF) membranes (Amersham). The membrane was then blocked with 5% non-fat dry milk in TBS and incubated with primary antibody (1:1000) overnight at 4 °C. Following incubation with HRP‐labelled secondary antibody (1:1000) at room temperature, the signal was detected using an ECL kit (Amersham) and visualized using Autoradiography.

Protein production

For protein production, HEK293T cells were transfected at 70% confluency in 6 × 10 cm plates. Cells were collected 48 h post-transfection and lysed using NP-40 lysis buffer (25 mM Tris-HCl pH 7.4, 150 mM NaCl, 1% NP-40, 5% glycerol) supplemented with protease cocktail inhibitor, and the protein-containing supernatant was collected. Spike protein was purified from the supernatant via affinity purification column and quantified using BCA assay and confirmed with western blot. Produced spike protein was stored at -20 °C in 50% glycerol and thawed upon usage.

Cell cytotoxicity using MTT assay

MTT assay was performed on Vero cells to evaluate the cytotoxicity of the selected compounds as previously described (Loutfy et al. 2022; Mosmann 1983). Briefly, 104 cells per well were seeded in 96-well plates in DMEM complete media. The following day, cells were treated with different concentrations of the compounds in triplicates. After 48 h, MTT dye was added to the cells (0.5 mg/mL) and incubated for 2 h. The plate was then decanted and 100 µL of DMSO was added per well. Absorbance readings were obtained at 570 nm using a microplate reader (CLARIOstar plus, BMG LabTech, Germany). The cell viability was then expressed as the percentage of the untreated control and CC50 (defined as the concentration that reduced cell viability by 50%) values of the tested compounds were calculated by GraphPad Prism 8 software.

Validation of spike protein binding activity

To validate the binding activity of the produced spike protein (SP) versus the commercial spike protein (SC) we allowed it to interact with ACE2 using our previously published protein-protein interaction detection method (Emam et al. 2022). Briefly, small glass slides of equal size, 1 cm x 1 cm x 1 mm, were cleaned, coated with APTES, and then activated with EDC-NHS. ACE2 protein was immobilized on the slides, 100 nM on each slide, then they were rinsed with PBS. Both SP and SC were diluted to 2 concentrations 10 nM and 100 nM. Each slide was then incubated with one of the spike proteins followed by the anti-spike antibody (10 nM), then the FITC-labelled secondary antibody (10 nM) with rinsing between each incubation. Two negative controls were included in each run to test for specificity. The negative control was subjected to all the mentioned steps excluding the addition of spike protein.

Fluorescence intensity readings were recorded before and after adding spike protein and after the final wash by transferring slides into 24-well plates and using a microplate reader (CLARIOstar plus, BMG LabTech, Germany) for signal detection.

Standardization of the in-house immunofluorescent assay for drug screening

Chitosan nanoparticles were used as a drug for the optimization of the assay conditions. CNPs preparation and characterization were previously described (Loutfy et al. 2022). The modified assay was performed following the same procedures as described in the previous section except for an extra incubation step between the immobilized protein (ACE2) and CNPs for 40 min, followed by rinsing with PBS. In the current assay, various concentrations of CNPs (1, 5, 10, 20, 30, and 50 µg/ml) were used (Emam et al. 2021).

Following standardization, we screened the selected compounds: Curcumin (Cur), Gallic acid (GA), Quercetin (Q), and Silymarin (Sil). Taking into consideration the CC50 values of each compound, different concentrations ranging from 5 µg/mL to 500 µg/mL were used. The IC50 of each compound, which represented 50% inhibition of the substrate (ACE2) binding to its ligand (or to the spike protein), was determined using GraphPad Prism 8 software.

In each run, positive and negative controls were performed. However, the controls were counterintuitive as the negative control included the addition of spike & ACE2 proteins and the antibodies, producing a high signal. While for the positive control, we excluded the addition of spike protein, subsequently, producing no signal.

Results

Production of SARS-CoV-2 spike protein

As an affordable alternative to the commercial spike protein (SC), we produced spike protein (SP) by transfecting HEK293T cells with spike ectodomain-expressing plasmid. The efficiency of transfection and spike protein expression was determined using western blot analysis. Major bands were observed in lanes A and B (transfected cells) (Fig. 1) at 180 kD corresponding to full-length SARS-CoV-2 spike protein. While no bands were observed in lane C showing the non-transfected negative control. Accordingly, we performed transfection on a larger scale and purified 920 µg of SP. (Fig. S1) shows the western blot original autoradiogram.

Fig. 1
figure 1

Spike protein expression. Expression of the spike protein was detected using western blot analysis after transfection. Lanes (A) and (B) represent samples from transfected HEK293T cells, showing bands at 180 kDa corresponding to the spike protein. Lane (C) represents the negative control

Validation of spike protein (SP) binding activity to ACE2

In order to compare the binding activity of both the produced SP and the SC to the ACE2 protein, we tested each at 2 concentrations: 10 and 100 nM via immunofluorescent assay. Both SP and SC produced similar signals when detected at each of the 2 concentrations (Fig. 2), proving that SP binding activity remained intact after the purification process.

Fig. 2
figure 2

Validation of SP binding activity as compared to SC. The fluorescent intensity readings were detected at 2 concentrations for each protein and plotted with Origin software version 2018

Determination of the cytotoxicity of the four natural compounds using MTT

CNPs, Cur, GA, Q, and Sil were tested and their CC50 values were detected at 175, 13, 18, 73, and 65 µg/mL, respectively (Table 1).

Table 1 CC50 values of the screened compounds on Vero cells

Standardization of in-house immunofluorescent assay for the determination of the inhibitory effect of selected compounds

As the ability of CNPs to inhibit spike-ACE2 interaction was previously identified (Loutfy et al. 2022), they were used in the standardization of the new immunofluorescent assay for drug screening. Different concentrations of CNPs were screened using the modified protocol of our protein-protein interaction detection assay (Emam et al. 2022). The signal was decreased after the rinsing of the labeled-secondary antibody step, in which the variation in fluorescence signals was linearly dependent on the concentration of the CNPs (Fig. 3A) proving the specificity of the observed binding.

This was further proved by the controls. The negative control (containing spike and ACE2) curve shows a high fluorescence intensity that corresponds to the binding between the ACE2 and the spike protein. In contrast, the positive control (containing ACE2 only) curve shows decreased fluorescence intensity which indicates the absence of spike-ACE2 interaction (Fig. 3B). Therefore, inhibition of spike-ACE2 interaction resembles the positive control here.

Fig. 3
figure 3

Standardization of the in-house immunofluorescent assay. (A) The effect of different CNPs concentrations on the signal intensity. The reduction in the signal corresponds to the increase in CNPs concentration, and (B) The negative and positive controls, the positive control (containing ACE2 only) shows no signal while the negative control (containing spike and ACE2) shows a high signal

Screening of four bioactive natural compounds using the standardized immunofluorescent assay

Results showed that the signal representing spike-ACE2 PPI was markedly hindered after treatment with any of the tested compounds. Hence the decrement of the signal corresponded to the increase in the compounds’ concentrations, indicating their ability to disrupt spike-ACE2 PPI. The compounds’ inhibitory effect was represented as IC50 values (Fig. 4). CNPs, Cur, GA, Q, and Sil IC50 values were determined at 12.69, 1.352, 4.943, 8.503, and 21.05 µg/mL, respectively.

Fig. 4
figure 4

Inhibitory effect of screened compounds. The drug-response curve was calculated for each drug with GraphPad Prism 8 software. IC50 values of CNPs, Cur, GA, Q, and Sil were detected at 12.69, 1.352, 4.943, 8.503, and 21.05 µg/mL

Discussion

The development of new laboratory methods that are simple enough to facilitate the drug discovery process is important and of high value, especially during pandemics when multiple research groups are working simultaneously and require affordable methods.

This study aimed to explore the applicability of our immunofluorescent protein-protein interaction (PPI) detection method for drug screening (Emam et al. 2022). Focusing on the well-known SARS-CoV-2 Spike protein and Angiotensin Converting Enzyme 2 (ACE2) interaction, we standardized the assay using chitosan nanoparticles (CNPs) as a known inhibitor (Loutfy et al. 2022). The successful screening of CNPs against Spike-ACE2 interaction allowed us to calculate the IC50 value, representing 50% inhibition of tested protein-protein interaction. We also sought to reduce screening costs by producing the spike protein (SP) in HEK293T cells, ensuring its binding activity to ACE2 was comparable to the commercial spike protein (SC).

It has been reported that some natural compounds are promising candidates for curing many diseases. Their significant pharmacological effects and mode of action as well as their low toxicity allowed them to be thought of as treatments for COVID-19 (Al-Harrasi et al. 2022; Bizzoca et al. 2022; Chakravarti et al. 2021; Khan et al. 2021). Therefore, After the standardization of the screening assay and spike protein production, we selected four natural compounds, Curcumin (Cur), Gallic acid (GA), Quercetin (Q), and Silymarin (Sil), as candidates to be screened using our assay. We assessed their cytotoxicity using MTT assay on Vero cells to obtain CC50 values, representing 50% reduction of the cell viability, before screening for their inhibitory activity.

Our results were in agreement with other reported results. Low and colleagues reported that concentrations of Silymarin up to 200 µg/mL did not show significant toxicity on Vero cells (Low et al. 2021). Furthermore, Ortiz-Andrade et al. evaluated the cytotoxic effect of Quercetin and reported its safety on Vero cells, and that only high concentrations ranging from 250 to 750 µg/mL decreased the cellular viability (Ortiz-Andrade et al. 2020). Additionally, Marin-Palma reported that CC50 of Curcumin on Vero E6 cells was obtained at 16.5 µg/mL (Marín-Palma et al. 2021). Moreover, Abdullah and colleagues determined the antiproliferative activity of Gallic acid to be 10.00 ± 1.06 µg/mL on HeLa cells (Abdullah et al. 2023).

After cytotoxicity evaluation of the selected natural compounds, they were screened using the standardized protocol for drug screening using our immunofluorescent assay. All the screened compounds were able to effectively disrupt the Spike-ACE2 interaction, with Curcumin demonstrating the strongest inhibitory effect, having the lowest IC50 value (1.352 µg/mL) which is even lower than that of our positive control, CNPs (12.69 µg/mL).

This is supported by previously reported data about the anti-viral effect of Curcumin against viruses, especially SARS-CoV-2. Curcumin demonstrated antiviral activity against influenza A virus, SARS-CoV, human immunodeficiency virus (HIV), enterovirus 71 (EV71), herpes simplex virus (HSV), hepatitis C virus (HCV), human papillomavirus (HPV), Zika virus, and Chikungunya virus; Multiple mechanisms contributed to its antiviral activity (Babaei, Nassiri-Asl, and Hosseinzadeh 2020; Soni et al. 2020). Furthermore, in-silico studies predicted the binding of curcumin with receptors that mediate viral entry of SARS-CoV-2 into host cells (Soni et al. 2020).

We can see that, this approach provides an easy and rapid screening tool for anti-SARS-CoV-2 candidates. However, the main limitation of our assay lies in its inapplicability to High Throughput Screening (HTS). This limitation arises from its reliance on glass slides that need to be transferred to a plate for microplate reader analysis, unlike ELISA assays that can be directly performed in a 96-well plate. Despite this limitation, our study contributes a valuable tool using cost-effective and efficient screening method for drugs under investigation.

In summary, our study successfully demonstrated the feasibility of utilizing our previously established immunofluorescent assay for drug screening against the Spike-ACE2 interaction. We produced a cost-effective in-house spike protein, which was highly comparable to the commercial one. The screening of natural compounds, particularly Curcumin, showed promising inhibitory effects with acceptable safety profiles. While the assay has limitations concerning HTS, its cost-effectiveness, and reproducibility make it a valuable tool for targeted drug screening in the context of specific protein-protein interaction. Further research is needed to confirm the antiviral effects in vitro and in vivo of the screened compounds, determining their potential as additional drugs for COVID-19 treatment.

References

  • Abdullah H, Ismail I, Suppian R, Nor Munirah Z (2023) Natural gallic acid and Methyl Gallate induces apoptosis in hela cells through regulation of intrinsic and extrinsic protein expression. Int J Mol Sci 24(10). https://doi.org/10.3390/ijms24108495

  • Ahamad S, Ali H, Secco I, Giacca M, Gupta D (2022) Anti-fungal drug Anidulafungin inhibits SARS-CoV-2 Spike-Induced Syncytia formation by Targeting ACE2-Spike Protein Interaction. Front Genet 13. https://doi.org/10.3389/fgene.2022.866474

  • Al-Harrasi A, Behl T, Upadhyay T, Chigurupati S, Bhatt S, Sehgal A, Bhatia S, Singh S, Sharma N, Vijayabalan S Vasanth Raj Palanimuthu, Suprava Das, Rajwinder Kaur, Lotfi Aleya, and Simona Bungau. 2022. ‘Targeting Natural products against SARS-CoV-2’. Environ Sci Pollut Res 29(28):42404–42432. https://doi.org/10.1007/s11356-022-19770-2

  • Assadiasl S, Fatahi Y, Mosharmovahed B, Mohebbi B, Mohammad Hossein Nicknam (2021) Baricitinib: from rheumatoid arthritis to COVID-19. J Clin Pharmacol 61(10):1274–1285. https://doi.org/10.1002/jcph.1874

    Article  PubMed  PubMed Central  Google Scholar 

  • Babaei F, Marjan Nassiri-Asl, and Hossein Hosseinzadeh (2020) Curcumin (a constituent of Turmeric): New Treatment option against COVID-19. Food Sci Nutr 8(10):5215–5227. https://doi.org/10.1002/fsn3.1858

    Article  PubMed  PubMed Central  Google Scholar 

  • Bizzoca M, Eleonora S, Leuci MD, Mignogna Eleonora Lo Muzio, Vito Carlo Alberto Caponio, and Lorenzo Lo Muzio. 2022. ‘Natural compounds may contribute in preventing SARS-CoV-2 infection: a narrative review’. Food Sci Hum Wellness 11(5):1134–1142. https://doi.org/10.1016/j.fshw.2022.04.005

  • Bojadzic D, Alcazar O, Chen J, Chuang ST, Jose M, Condor Capcha LA, Shehadeh, and Peter Buchwald (2021) Small-molecule inhibitors of the Coronavirus Spike: ACE2 protein-protein Interaction as blockers of viral attachment and entry for SARS-CoV-2. ACS Infect Dis 7(6):1519–1534. https://doi.org/10.1021/acsinfecdis.1c00070

    Article  PubMed  PubMed Central  Google Scholar 

  • Bozorgmanesh M, Yousefifar Y, Jamalian M, Mahmoodiyeh B, Kamali A (2021) A review on common treatments for COVID-19 and Use of Tocilizumab (ACTEMRA) in reducing effects of the Disease. Annals Romanian Soc Cell Biology 25(4):2920–2936

    Google Scholar 

  • Chakravarti R, Singh R, Ghosh A, Dey D, Sharma P, Velayutham R, Roy S, Dipanjan Ghosh (2021) A review on potential of Natural products in the management of COVID-19. RSC Adv 11(27):16711–16735. https://doi.org/10.1039/d1ra00644d

    Article  PubMed  PubMed Central  Google Scholar 

  • Emam MH, Ahmed I, Abdel-Salam Y, Moatasim MR, Gomaa, Nasra F, Abdel Fattah F, Ali HM, Alam El-Din A, Mostafa MA, Ali SA, Loutfy (2021) and Amal Kasry. ‘Development of a Newly Confirmatory Immunoassay to Evaluate Anti-Viral Activity of Chitosan Nanoparticles (Cnps) against SARS-CoV-2 Spike Protein’. in The annual conference of National Cancer Institute - Cairo University ‘Bridging Gaps in Oncology’. Cairo: Journal of the Egyptian National Cancer Institute

  • Emam MH, Nageh H, Ali F, Taha M, ElShehaby HA, Amin R, Kamoun EA, Loutfy SA, and Amal Kasry (2022) Inhibition of SARS-CoV-2 spike protein entry using biologically modified Polyacrylonitrile nanofibers: in Vitro Study towards specific antiviral masks. RSC Adv 12(25):16184–16193. https://doi.org/10.1039/d2ra01321e

    Article  PubMed  PubMed Central  Google Scholar 

  • Govea-Salas M, Rivas-Estilla AM, Rodríguez-Herrera R, Lozano-Sepúlveda SA, Aguilar-Gonzalez CN, Zugasti-Cruz A, Salas-Villalobos TB, Jesus Antonio Morlett-Chávez (2016) Gallic acid decreases Hepatitis C Virus expression through its antioxidant capacity. Experimental Therapeutic Med 11(2):619–624. https://doi.org/10.3892/etm.2015.2923

    Article  Google Scholar 

  • Hiremath, Shridhar HD, Vinay Kumar M, Nandan M, Mantesh KS, Shankarappa V, Venkataravanappa CR, Jahir, Basha, Lakshminarayana CN (2021) Reddy. ‘In Silico Docking Analysis Revealed the Potential of Phytochemicals Present in Phyllanthus Amarus and Andrographis Paniculata, Used in Ayurveda Medicine in Inhibiting SARS-CoV-2’. 3 Biotech 11(2). https://doi.org/10.1007/s13205-020-02578-7

  • Jadel, Müller, Kratz Carla Regina Andrighetti-Fröhner, Paulo César Leal, Ricardo José Nunes, Rosendo Augusto Yunes, Edward Trybala, Tomas Bergström, Célia Regina Monte Barardi, and Cláudia Maria Oliveira Simões. 2008. ‘Evaluation of Anti-HSV-2 activity of gallic acid and Pentyl Gallate’. Biol Pharm Bull 31(5):903–907. https://doi.org/10.1248/bpb.31.903

  • Jia H, Neptune E, Cui H (2021) Targeting ACE2 for COVID-19 therapy: opportunities and challenges. Am J Respir Cell Mol Biol 64(4):416–425. https://doi.org/10.1165/rcmb.2020-0322PS

    Article  PubMed  PubMed Central  Google Scholar 

  • Khan N, Chen X, Geiger JD (2021) Possible therapeutic use of natural compounds against COVID-19. J Cell Signal 2(1):63–79

    PubMed  PubMed Central  Google Scholar 

  • Lai C-C, Po‐Ren H (2023) Coronavirus Disease 2019 Rebounds following Nirmatrelvir/Ritonavir Treatment. J Med Virol 95(2). https://doi.org/10.1002/jmv.28430

  • Lamb YN (2020) Remdesivir: first approval. Drugs 80(13):1355–1363. https://doi.org/10.1007/s40265-020-01378-w

    Article  PubMed  PubMed Central  Google Scholar 

  • Li Y, Yao J, Han C, Yang J, Chaudhry MT, Wang S, Liu H, Yulong Yin (2016) Quercetin, inflammation and immunity. Nutrients 8(3). https://doi.org/10.3390/nu8030167

  • Loutfy SA, Ahmed I, Abdel-Salam Y, Moatasim MR, Gomaa, Nasra F, Abdel Fattah MH, Emam F, Ali HA, ElShehaby, Eman A, Ragab HM. Alam El-Din, Ahmed Mostafa, Ali MA, and Amal Kasry (2022b). ‘Antiviral Activity of Chitosan Nanoparticles Encapsulating Silymarin (Sil-CNPs) against SARS-CoV-2 (in Silicoin Vitro Study)’. RSC Advances 12(25):15775–86. https://doi.org/10.1039/d2ra00905f

  • Low Z, Xuan BM, OuYong P, Hassandarvish CL, Poh, and Babu Ramanathan (2021) Antiviral activity of silymarin and Baicalein against Dengue Virus. Sci Rep 11(1). https://doi.org/10.1038/s41598-021-98949-y

  • Marín-Palma D, Tabares-Guevara JH, Zapata-Cardona MaríaI, Flórez-álvarez L, Yepes LM, Maria T, Rugeles JC, Hernandez (2021) and Natalia A. Taborda. ‘Curcumin Inhibits in Vitro Sars-Cov-2 Infection in Vero E6 Cells through Multiple Antiviral Mechanisms’. Molecules 26(22). https://doi.org/10.3390/molecules26226900

  • Mosmann T (1983) Rapid Colorimetric Assay for Cellular Growth and Survival: Application to Proliferation and Cytotoxicity Assays. Vol. 65

  • Murakami N, Hayden R, Hills T, Al-Samkari H, Casey J, Sorbo LD, Lawler PR, Sise ME (2023) and David E. Leaf. ‘Therapeutic Advances in COVID-19’. Nature Reviews Nephrology 19(1):38–52. https://doi.org/10.1038/s41581-022-00642-4

  • Ortiz-Andrade R, Jesús Alfredo Araujo-León, Amanda Sánchez-Recillas, Gabriel Navarrete-Vazquez, Avel Adolfo González-Sánchez, Sergio Hidalgo-Figueroa, Ángel Josabad Alonso-Castro, Irma Aranda-González, Emanuel Hernández-Núñez, Tania Isolina Coral-Martínez, Juan Carlos Sánchez-Salgado, Victor Yáñez-Pérez, and, Lucio-Garcia MA (2020) ‘Toxicological Screening of Four Bioactive Citroflavonoids: In Vitro, In Vivo, and In Silico Approaches’. Molecules 25(24). https://doi.org/10.3390/MOLECULES25245959

  • Pan B, Fang S, Zhang J, Pan Y, Liu H, Wang Y, Li M, Liren Liu (2020) Chinese Herbal compounds against SARS-CoV-2: Puerarin and quercetin impair the binding of viral S-Protein to ACE2 receptor. Comput Struct Biotechnol J 18:3518–3527. https://doi.org/10.1016/j.csbj.2020.11.010

    Article  PubMed  PubMed Central  Google Scholar 

  • Rattis BAC, Simone G, Ramos, Celes MRN (2021) Curcumin as a potential treatment for COVID-19. Front Pharmacol 12. https://doi.org/10.3389/fphar.2021.675287

  • Saravolatz LD, Depcinski S, Sharma M (2023) Molnupiravir and Nirmatrelvir-Ritonavir: oral coronavirus Disease 2019 antiviral drugs. Clin Infect Dis 76(1):165–171. https://doi.org/10.1093/cid/ciac180

    Article  PubMed  Google Scholar 

  • Scialo F, Daniele A, Amato F, Pastore L, Matera MG, Cazzola M, Castaldo G, Bianco A (2020) ACE2: the major cell entry receptor for SARS-CoV-2. Lung 198(6):867–877. https://doi.org/10.1007/s00408-020-00408-4

    Article  PubMed  PubMed Central  Google Scholar 

  • Soni V, Kumar A, Mehta YK, Ratre AK, Tiwari A, Amit RP, Singh Subash Chandra Sonkar, Navaneet Chaturvedi, Dhananjay Shukla, and Naveen Kumar Vishvakarma. 2020. ‘Curcumin, a traditional spice component, can hold the Promise against COVID-19?’. Eur J Pharmacol 886. https://doi.org/10.1016/j.ejphar.2020.173551

  • Tedesco F, Calugi L, Lenci E, Trabocchi A (2022) Peptidomimetic Small-Molecule inhibitors of 3CLPro Activity and Spike-ACE2 Interaction: toward dual-action molecules against Coronavirus infections. J Org Chem 87(18):12041–12051. https://doi.org/10.1021/acs.joc.2c01047

    Article  PubMed  PubMed Central  Google Scholar 

  • Vahedian-Azimi A, Abbasifard M, Rahimi-Bashar F, Guest PC, Majeed M, Mohammadi A, Banach M Tannaz Jamialahmadi, and Amirhossein Sahebkar. 2022. ‘Effectiveness of Curcumin on outcomes of hospitalized COVID-19 patients: a systematic review of clinical trials’. Nutrients 14(2). https://doi.org/10.3390/nu14020256

  • Wrapp D, Wang N, Corbett KS, Goldsmith JA, Hsieh C-L, Abiona O, Graham BS, Mclellan JS (2019) Cryo-EM Structure of the 2019-NCoV Spike in the Prefusion Conformation

  • Xu D, Wang MJHYQ, Yuan Lu C (2019) Antioxidant activities of Quercetin and its complexes for Medicinal Application. Molecules 24(6). https://doi.org/10.3390/molecules24061123

Download references

Funding

Open access funding provided by The Science, Technology & Innovation Funding Authority (STDF) in cooperation with The Egyptian Knowledge Bank (EKB).

Author information

Authors and Affiliations

Authors

Contributions

M.H.E performed the cytotoxicity in-vitro assays, as well as the immunofluorescent screening assay, conducted the graphical analysis for the results, and initiated the writing of the original draft. M.I.M produced and purified the in-house spike protein, conducted the graphical analysis for the results, and participated in the writing of the manuscript. All experiments were conducted under the supervision and guidance of S.A.L, and N.E. S.A.L, and N.E reviewed and supervised the writing of the manuscript. S.A.L was also responsible for the development of the main idea, managing the resources, and funding acquisition. All authors revised the manuscript and agreed to all its contents.

Corresponding author

Correspondence to Samah A. Loutfy.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

All authors agree to be published.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Emam, M.H., Mahmoud, M.I., El-Guendy, N. et al. Establishment of in-house assay for screening of anti-SARS-CoV-2 protein inhibitors. AMB Expr 14, 104 (2024). https://doi.org/10.1186/s13568-024-01739-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13568-024-01739-8

Keywords