Skip to main content

Dual action of epigallocatechin-3-gallate in virus-induced cell Injury

Abstract

Background

Viral infections cause damage and long-term injury to infected human tissues, demanding therapy with antiviral and wound healing medications. Consequently, safe phytochemical molecules that may control viral infections with an ability to provide wound healing to viral-induced tissue injuries, either topically or systemically, are advantageous. Herein, we hypothesized that epigallocatechin-3-gallate (EGCG), the most abundant polyphenol in green tea, might be effective as a wound healing, antiviral, and antifibrotic therapy.

Results

The antiviral activities of EGCG against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and Herpes simplex virus type 2 (HSV-2) as well as its wound healing activities against different monolayer tissue (continuous and primary) systems were investigated. Consider its possible wound-healing advantages as well. To determine the safe concentrations of EGCG in green monkey kidney (Vero) and Vero-E6 cell lines, MTT assay was performed and showed high CC50 values of 405.1 and 322.9 μM, respectively. The antiviral activities of EGCG against SARS-CoV-2 and HSV-2, measured as half-maximal concentration 50 (IC50) concentrations, were 36.28 and 59.88 μM, respectively. These results confirm that the EGCG has remarkable viral inhibitory activities and could successfully suppress the replication of SARS-CoV-2 and HSV-2 in vitro with acceptable selectivity indices (SI) of 11.16 and 5.39, respectively. In parallel, the EGCG exhibits significant and dose/time-dependent anti-migration effects in human breast cancer cells (MCF-7), its resistant variation (MCF-7adr), and human skin fibroblast (HSF) indicating their potential to heal injuries in different internal and topical mammalian systems.

Conclusions

The EGCG has proven to be an efficient antiviral against SARS-CoV-2 and HSV-2, as well as a wound-healing phytochemical. We assume that EGCG may be a promising option for slowing the course of acute cellular damage induced by systemic (Coronavirus Disease 2019 (COVID-19)) or topical (HSV-2) viral infections.

Background

Antiviral therapy is the first control option to consider once a virus infection is established. All the steps in the virus life cycle ranging from entry to release can be explored as molecular targets for specific antiviral therapy as well as virus-dependent cell targets for indirect antiviral therapy. Moreover, antiviral therapy should certainly be used if patients are at high risk for tissue injury via virus-induced cellular responses or if patients are immunocompromised.

SARS-CoV-2 is one of many viruses that infect the human respiratory system, causing symptoms ranging from modest upper airway involvement to life-threatening acute respiratory distress syndrome (ARDS). Lung consequences, as illustrated by the current COVID-19 pandemic, include pneumonia and acute respiratory distress syndrome (ARDS) in critical cases [1, 24, 52]. Due to the combined effect of direct viral and indirect patient-specific immune-mediated damage, the clinical picture of lung tissue destruction is difficult to anticipate. It is widely known that SARS-CoV-2 interacts with the angiotensin-converting enzyme 2 (ACE2), which is predominantly found in type II pneumocytes in the lungs. ACE2 expression changes as a result of viral binding appear to be associated with increased vascular permeability, increased lung edema, increased lung injury, and overproduction of proinflammatory factors [16, 29]. In clinical terms, the lungs become irritated and filled with fluid, resulting in breathing difficulties. Breathing issues in certain individuals can become serious enough to require hospitalization with an oxygen ventilator. Consequently, lung injury may take months to improve. Another potential COVID-19 consequence is sepsis, which occurs when an infection spreads through the circulatory system, causing tissue damage everywhere it goes. Sepsis, even if a patient survives, can cause long-term damage to the lungs and other organs [18,19,20].

On the other hand, Herpes simplex virus (HSV) is another prevalent viral disease globally. HSV type-1 (HSV-1) infection occurs in or around the mouth and is mostly spread by oral-to-oral contact, whereas HSV type-2 (HSV-2) infection occurs sexually and causes genital herpes [42]. Both oral and genital herpes are usually asymptomatic. Nevertheless, they can cause painful blisters or ulcers, ranging from moderate to severe [4]. Although herpesviruses do not primarily target the lungs,under certain circumstances, several of them can cause interstitial pneumonia, bronchopneumonia, and ARDS. Infected persons commonly report tingling, stinging, or burning around their lips prior to the onset of sores. These symptoms may return regularly, with the frequency changing depending on the individual [32]. Besides, HSV infects the lower respiratory tract (LRT) of immunocompetent and immunocompromised patients [11, 9]. For instance, in many burned or intubated patients with squamous metaplasia of the respiratory epithelium, the spreading of the virus to the lung is probably an extension or aspiration of oropharyngeal HSV, or via hematogenous spread [11].

In any case, the rising trend in the prevalence of viral diseases drives researchers to explore treatment solutions that not only inactivate the virus but also aid in the process of regenerating wounded cells in order to mitigate the cellular damaging consequences of viral infection.

Natural flavonoids, a diverse set of polyphenolic substances found in plants, should be explored as a potential treatment for viral infections. For instance, the polyphenol epigallocatechin-3-gallate (EGCG), a major active ingredient in green tea, has been shown to have anti-inflammatory [35], antioxidant [10], anti-fibrotic [37], antimicrobial [53] and antiviral activities [51]. In this study, we investigated the possible use of EGCG to control viral infections with RNA and DNA model viruses of known viral-induced cell injuries. The antiviral activity against pandemic SARS-CoV-2 and HSV-2 and the possible wound-healing activity following the induction of cell-monolayer injuries in various mammalian systems were studied.

Results

The cytotoxicity (CC50) of EGCG on Vero and Vero-E6 cell lines

The EGCG was serially two-fold diluted and added to the cell culture medium to examine its effect on the growth and viability of Vero and Vero-E6 cell lines. After 3 days of co-incubation, the cell viability of Vero and Vero–E6 cells was determined using MTT assay. The mean dose–response curve of three different experiments was used to calculate the 50% cytotoxic and growth inhibition doses. The half-maximal cytotoxic concentrations of EGCG was determined to be 322.9 and 405.1 μM in Vero and Vero-E6 cells, respectively (Fig. 1).

Fig. 1
figure 1

Cytotoxicity and antiviral activity of EGCG against SARS-CoV-2 (a) and HSV-2 (b). The Cytotoxicity and antiviral activity were determined in Vero E6 and Vero cells against NRC-03-nhCoV and HSV-2, respectively. Half maximal cytotoxic (CC50) and inhibitory (IC50) concentrations were calculated using nonlinear regression analysis of GraphPad Prism software (version 5.01) by plotting log inhibitor versus normalized response (variable slope)

EGCG exerts antiviral activity against SARS-CoV-2 and HSV-2 in vitro

The cytopathic inhibition experiment was used to investigate EGCG's antiviral activity against SARS-CoV-2 and HSV-2. As a control, untreated virus-infected cells were employed in the test. The 50% inhibitory concentration (IC50) values for SARS-CoV-2 and HSV-2 were determined to be 36.28 and 59.88 μM, respectively (Fig. 1).

The selective indices are calculated by dividing the CC50 by IC50 (SI = CC50/IC50) values and are found to be 11.16 and 5.39 for SARS-CoV-2 and HSV-2, respectively (Table 1). At non-cytotoxic EGCG concentrations, SARS-CoV-2 infectivity was reduced by more than 59%, while HSV-2 infectivity was decreased by 41%.

Table 1 Inhibitory Concentration (IC50) and selective index (SI) for EGCG against both viral infections

Stage of antiviral activity

To confirm whether the anti-SARS-CoV-2 and anti-HSV-2 activity of EGCG can be attributed to the inhibition of the virus in a cell-free status or virus adhesion to the host cell receptors or replication inside the host cell, plaque reduction assays were performed as previously described [14]. Consequently, the EGCG affected both viruses mainly by targeting them in a cell-free status/virucidal effect (upto 100% inhibition of SARS-CoV-2 virus and 69.97 ± 7.1% of HSV-2), followed by interference with viral adsorption (average viral inhibition equals 80.1 ± 5.3% for SARS-CoV-2 and 35.1 ± 6.21 for HSV-2). The interference of EGCG with the replication efficiency of both viruses was low (average viral inhibition equals 17.44 ± 8.5% for SARS-CoV-2 and 15.03 ± 2.2% for HSV-2), when compared to the other two mechanisms or replication-cycle stages (Fig. 2 a & b).

Fig. 2
figure 2

Stage of antiviral activity of EGCG. The EGCG (50 μM) has been tested against SARS-CoV-2 (a) and HSV-2 (b) at different stages of the viral replication cycle including cell-free status “virucidal effect”, adsorption inhibition, and interference with viral replication

EGCG exerts an anti-migration effect against MCF-7 and its resistant variant (MCF-7 adr ) cells

The anti-migration effects of EGCG against MCF-7 and its resistant variant MCF-7adr were assessed in-vitro using a wound healing assay to reflect the potential of EGCG in suppressing tumor invasion. MCF-7 and MCF-7adr cells were treated with 1 μM and 10 μM of EGCG for 24 h (Day-1), 48 h (Day-2), and 72 h (Day-3), and wound closure was compared to the control untreated group. In MCF-7 cells, after 24 h treatment with EGCG, no significant change in wound gap was observed between treated and control cells. Further exposure of cells to EGCG significantly delayed wound closure compared to control untreated cells. After 48 h, EGCG (10 μM) showed a significant migration inhibition in MCF-7 cells (wound gap of 263.7 ± 9.3 μm) compared to control untreated cells with wound gap of 227.9 ± 17.0 μm. Prolonged exposure of cells to both 1 μM and 10 μM of EGCG for 72 h significantly delayed the cell migration with a wound gap of 156.3 ± 12.6 μm-wide and 202.9 ± 37.6 μm-wide, respectively, while the gap in control untreated cells was 122.3 ± 15.2 μm (Fig. 3 a & b).

Fig. 3
figure 3

The effect of EGCG on MCF-7 and MCF-7adr cell migration. MCF-7 and MCF-7adr cell monolayer sheets were scratched and treated with EGCG (1 and 10 μM). Images for the wound gap distance were taken for treated and control cells after 24 h, 48 h, and 72 h (a & c). Wound gap distances were optically measured and displayed over time (b & d). Data are shown as mean ± SD; n = 3. *P < 0.05, versus the control group

In MCF-7adr cells, EGCG significantly delayed wound closure compared to untreated control cells as early as after 24 h exposure. Wound gap distances were found to be 197.6 ± 38.5 μm-wide and 186.3 ± 17.1 μm-wide after treatment with 1 μM and 10 μM of EGCG, respectively, compared to wound gap of 138.6 ± 23.4 μm for untreated control cells. After 48 h, EGCG (1 μM 10 μM) showed further significant migration inhibition with a wound gap of 94.6 ± 13.9 μm-wide and 108.6 ± 26.9 μm-wide, respectively, while the gap in control untreated cells was 41.6 ± 29.4 μm (Fig. 3 c & d). Therefore, it can be concluded that EGCG exhibited a significant dose/time-dependent anti-migration effect against MCF-7 cells and its resistant variant (MCF-7adr).

EGCG facilitates wound healing effect using normal human skin fibroblast cells

Another common interpretation for the scratch assay is the wound healing capacity when the drug under investigation is tested against normal skin fibroblast cells. Herein, we investigated the wound healing capacity of EGCG (1 and 10 μM) against HSF cells (human skin fibroblast). EGCG (10 μM) enhanced the wound closure as early as after 24 h with a wound gap of 119.7 ± 3.3 μm compared to 130.0 ± 24 μm for untreated control fibroblast. After 48 h, both concentrations of EGCG (1 μM and 10 μM) showed a total wound closure compared to a remaining wound gap of 48.3 ± 9.8 μm in the untreated control fibroblast cells (Fig. 4 a & b). Therefore, and in contrast to cancer cells, EGCG facilitates wound healing in normal fibroblast cells in a dose and time-dependent manner.

Fig. 4
figure 4

The effect of EGCG on HSF cell migration. HSF cell monolayer sheets were scratched and treated with EGCG (1 and 10 μM). Images for the wound gap distance were taken for treated and control cells after 24 h, 48 h, and 72 h (a). Wound gap distances were optically measured and displayed over time (b). Data are shown as mean ± SD; n = 3. *P < 0.05, versus the control group

Discussion

Acute systemic and topical viral infections including COVID-19 and herpes are usually associated with cell injuries and wounds that persist for longer times after termination of viral replication and shedding [34]. In addition, the COVID-19 pandemic is meaningfully associated with many social factors including depression that affect immune fitness [21], delaying the wound healing process [3, 40].

Since the emergence of COVID-19, the interest is growing among researchers to develop effective antivirals to alleviate disease progression, reduce viral replication and infection spreading. Nevertheless, the pathological picture of COVID-19 is usually associated with diffuse alveolar damage (DAD) that demand precise and immediate healing [39]. The abnormal wound healing of this damage may result in additional severe scarring and fibrosis than other forms of the life-threatening ARDS [30]. The stepwise recovery of this damage demands a dynamic innate and acquired immune responses and epithelial cells regeneration and induction via wound-healing phytochemicals rather than administrating epithelial growth factors that are more likely to be detrimental and could increase the viral load via the upregulation of the ACE2 expression on the host cells [30].

On the same hand, Herpes simplex virus is capable of causing topical partial-thickness wounds [38]. Similarly to COVID-19, herpes simplex virus infection delays healing of oral excisional and extraction wounds in experimental animals [13]. To these points, we sought to identify a safe phytochemical molecule that can serve as antiviral medication with the ability to aid in healing the infection-mediated epithelial/endothelial injury.

Epigallocatechin-3-gallate (EGCG), a form of catechin, is a well-known natural antiviral drug with the potential to inhibit numerous viruses as well as reduce oxidative damage and promote lung regeneration capacity [27, 31, 33, 45]. The EGCG has an inhibitory impact on a wide variety of pathogens and functions as a broad-spectrum antiviral agent. It is most common in the following locations: (a) EGCG inhibits cytomegalovirus [22], Zika virus [5], and human immunodeficiency virus [12, 26] aggressively during early infection,(b EGCG inhibits viral replication, including influenza virus [23], enterovirus [15], and hepatitis virus [57],and (c EGCG inhibits pathogens indirectly by regulating immune inflammation and oxidative stress [55].

Herein, the wound healing and antiviral effects of EGCG on multiple cell lines, as well as two viral pathogens, COVID-19 and HSV-2, were accordingly investigated. In parallel, EGCG showed remarkable antiviral efficacy against SARS-CoV-2 (RNA virus) and HSV-2 (DNA virus) at safe concentrations. The data in this study confirmed that EGCG has high CC50 values reflecting its low cytotoxicity. EGCG has low inhibitory concentration 50 (IC50) against SARS-CoV-2 and HSV-2 (36.28 and 59.88 μM, respectively), indicating its remarkable antiviral activity against RNA and DNA model viruses. The investigation of the stage at which EGCG can impair viral replication cycle, it was found in this study that it affects the viral particle directly “virucidal effect” as well as interferes with the virus ability to adsorb to the host cell receptors. Consistently, this study confirms that EGCG exhibits broad antiviral activity against numerous viruses [25, 28, 36, 49, 54]. It has previously been shown to prevent the attachment of hepatitis viruses, influenza, reovirus, coronavirus and vesicular stomatitis virus by acting directly on the virions or interacting with surface proteins [8, 25, 58]. These prior studies back up our findings and indicate that EGCG inhibits the propagation of a variety of viruses, including RNA and DNA viruses. EGCG has also been proposed to inhibit 3CL-Protease of SARS-CoV-2 [17]. The IC50 value as an indicator of the viral inhibitory effect was found to be variable against different viruses including SARS-CoV-2 [28]. This shows that EGCG can hinder the infectivity of different viral pathogens, however the wide disparity in IC50 values could be due to a number of factors such as EGCG extraction technique, cellular and viral model variations, or antiviral methods used.

Furthermore, EGCG has been shown and confirmed to have a wide anti-lung fibrosis impact. To the best of our knowledge, there is no reported relationship between COVID-19 induced lung fibrosis and type of lung cancers [6, 50]. Pulmonary fibrosis, for instance, develops in COVID-19 patients because SARS-CoV-2 infection induces a massive increase in neutrophil infiltration into the lungs, leading to TGF production. An unregulated surge in active TGF-beta 1, aided by proinflammatory cytokines such as TNF, IL-6, and IL-1, induces fast and widespread edema and fibrosis and eventually clogs the airways resulting in lung functional failure [6]. Some studies investigated the protective impact of EGCG against lung fibrosis and found that it enhanced lysosomal hydrolases and ultrastructural changes in the lungs of a bleomycin-induced rat model of lung fibrosis [46,47,48]. Furthermore, downregulating TGF-β1 signaling inhibited fibroblast activation and collagen buildup, providing solid evidence that EGCG is an effective anti-fibrotic medication [6]. In line with previous findings, the documented antioxidant, anti-inflammatory, antimicrobial, angiogenesis and antifibrotic properties of the EGCG potentiate its activity at diverse stages of topical wound healing including hemostasis, inflammation, proliferation and tissue remodeling [56, 59].

Over and above, EGCG is known to possess potential chemopreventive as well as chemotherapeutic actions [7]. In the current study, EGCG showed significant antiproliferative as well as anti-invasive properties in both naïve (MCF-7) and resistant (MCF-7Adr) tumor cells. Breast adenocarcinoma cells were used herein as a proof of principle and to compare the potential effect of EGCG on sister naïve and resistant cell lines.

Conclusions

To sum up, the potential inhibitory impacts of EGCG as an antiviral therapy option against SARS-CoV-2 and HSV-2 were confirmed in this study. In addition to its antiviral characteristics, EGCG has demonstrated potent and dose/time-dependent anti-invasion effects on malignant cell lines. On the top of these therapeutic effects, EGCG showed enhancement in wound healing properties on normal fibroblast cells which can cushion all previously mentioned effects’ collateral damages. This emphasizes that the EGCG can be further investigated in vivo in preclinical studies as a trial to be applied as a candidate potentially safe antiviral and wound-healing phytochemical in COVID-19 and HSV-2 infections.

Methods

Cell Culture, EGCG, and Viruses

Nawah Scientific Inc. (Mokattam, Cairo, Egypt) provided the African green monkey kidney (Vero), (Vero-E6), human breast cancer (MCF-7), doxorubicin-resistant breast cancer (MCF-7Adr), and human normal skin fibroblast (HSF) cell lines, which were cultured in DMEM (Dulbecco's Modified Eagle's Medium), Gibco, USA. The culture media was supplemented with 10% fetal bovine serum (FBS) and 100 units/mL penicillin/streptomycin (PS). The cells were incubated at 37 oC in a humid atmosphere with 5% CO2. Sigma-Aldrich (Seelze, Germany) provided the EGCG, which was diluted to stock solutions with PBS and kept at 80° C for all subsequent studies.

Nawah-Scientific Co. for Scientific Research Services, Egypt generously contributed HSV-2 for in vitro viral challenge. The hCoV-19/Egypt/NRC-3/2020 SARS-CoV-2 (NRC-03-nhCoV) was obtained from the virus collections of the Centre of Scientific Excellence for Influenza viruses at the National Research Centre, Egypt. The viral titers were calculated using the limit-dilution method and were expressed as a 50% cell culture infective dosage (TCID50) of 1 × 104 (SARS-CoV-2), and 1 × 106 (HSV-2), respectively. Virus stocks were kept at 80 °C until they were used.

Cytotoxicity of EGCG on Vero and Vero-E6 Cell lines

Based on prior reports [41, 43], cells were seeded in a 96-well plate at a density of 2 × 105 cells/well and then treated for 72 h at 37 °C in a humidified environment of 5% CO2 with two-fold concentrations of EGCG (7.5–0.0146 mM). Following the incubation period, the medium was replaced with 100 μl of MTT solution (5 mg/ml) and incubated at 37 °C for 4 h. After 30 min at 37 °C, the MTT solution was changed with 50 μl of acidified isopropanol. The absorbance at 570 nm was then measured to estimate the maximum concentration of EGCG that was not toxic to the cells using the next equation. (A-B/A) × 100 was used to calculate the 50% cytotoxic concentration (CC50), where A & B are the means of three OD570 measurements of untreated and treated cells, respectively.

Efficacy of EGCG against SARS-CoV-2 and, HSV-2 challenges in vitro

To investigate the antiviral activity of EGCG against SARS-CoV-2 and HSV-2, the half maximal inhibitory concentration 50 (IC50) were estimated as previously described [14]. Briefly, Vero and Vero-2 confluent 96-well plates were infected for 60 min at 37 oC with 100 μl of stock SARS-CoV-2, and HSV-2 viruses. The EGCG was then added in 100 μl increments. Three wells were utilized for each dilution, and 100 μl of the maintenance medium was added to each well. Plates were finally incubated for three days until cytopathic effect (CPE) was observed. Subsequently, the cells were fixed with 100 μL/well 10% fixing solution and incubated for 2 h at room temperature, then the supernatants were discarded and 50 μl/well of 0.1% crystal violet stain were added for 10 min. The dried stained treated and control wells were then supplemented with 180 μl/well of absolute methanol and shaked for 30 min. The optical density (OD) was then measured at 570 nm using ELISA plate reader. A plot of cell viability (%) and viral inhibition (%) versus concentration for each EGCG was represented using GraphPad prism 5 software.

Mode/Stage of antiviral action

To define the stage at which EGCG is affecting SARS-CoV-2 and HSV-2, three main stages of the viral replication cycle including (a) virucidal effect; (b) adsorption inhibition; and (c) replication interference, were investigated by plaque reduction assay of action as previously described [14]. The EGCG was applied in the three protocols at an effective concentration of 100 μM (< CC50 and > IC50 values). The percent of viral reduction was calculated using the following equation:

$$\mathbf{Plaque}\;\mathbf{reduction}\;(\boldsymbol{\%})=\frac{\mathbf{Count}\;\mathbf{of}\;\mathbf{untreated}\;\mathbf{virus}\;(\mathbf{control})-\mathbf{Count}\;\mathbf{of}\;\mathbf{treated}\;\mathbf{virus}}{\mathbf{Count}\;\mathbf{of}\;\mathbf{untreated}\;\mathbf{virus}\;(\mathbf{control})}\times100$$

Wound healing assay

Wound healing assay (scratch assay) was used herein to assess the anti-migration effect of EGCG against MCF-7 (human breast adenocarcinoma cells), and its resistant variant MCF-7adr and HSF (human skin fibroblast) cells. Briefly, 1 × 105 cells were seeded in a 6-well plate and maintained until a minimum of 80% confluent monolayer cell sheet. Afterward, monolayer cell sheets were scratched with a sterile pipette tip to generate a 40 μm-wide wound, and scratched cell debris was washed out with PBS. Fresh media or media containing EGCG were added to cells and incubated for a further 72 h. Wound closure was monitored every day by collecting digitized images using TCM-400 inverted microscope (LaboMed, Fremont, CA, USA) coupled with a Digital Still Camera (35 mm SLR camera) for scratch width calculation [2].

Statistical analysis

All experiments were performed in triplicate and calculations were carried out using GraphPad PRISM and linear regression analysis (Version 8.0.1, GraphPad Software, San Diego, CA, USA). The selective index (SI) was derived using CC50/IC50 [44].

Availability of data and materials

All data generated or analyzed during this study are included in this article.

References

  1. Al-Karmalawy AA, Soltane R, Abo Elmaaty A, Tantawy MA, Antar SA, Yahya G, Chrouda A, Pashameah RA, Mustafa M, Abu Mraheil M, Mostafa A (2021) Coronavirus Disease (COVID-19) Control between Drug Repurposing and Vaccination: A Comprehensive Overview. Vaccines (Basel). 9(11):1317

    Article  Google Scholar 

  2. Alqarni AA, Alamoudi AA, Allam RM, Ajabnoor GM, Harakeh SM, Al-Abd AM (2022) The influence of antioxidant dietary-derived polyphenolic combination on breast cancer: Molecular study. Biomed Pharmacother 149:112835

    Article  Google Scholar 

  3. Balikji J, Kiani P, Hendriksen PA, Hoogbergen MM, Garssen J, Verster JC (2022) Impaired wound healing is associated with poorer mood and reduced perceived immune fitness during the COVID-19 pandemic: A retrospective survey. Health Sci Rep 5:e764

    Article  Google Scholar 

  4. Cantatore A, Randall SD, Traum D, Adams SD (2013) Effect of black tea extract on herpes simplex virus-1 infection of cultured cells. BMC Complement Altern Med 13:139

    Article  Google Scholar 

  5. Carneiro BM, Batista MN, Braga ACS, Nogueira ML, Rahal P (2016) The green tea molecule EGCG inhibits Zika virus entry. Virology 496:215–218

    Article  Google Scholar 

  6. Chapman HA, Wei Y, Montas G, Leong D, Golden JA, Trinh BN, Wolters PJ, LeSaux CJ, Jones KD, HillsFosterOldhamLinderholmKotakDecarisTurnerSong NKEJMALPMSJW (2020) Reversal of TGFβ1-Driven Profibrotic State in Patients with Pulmonary Fibrosis. N Engl J Med 382:1068–1070

    Article  Google Scholar 

  7. Chen D, Wan SB, Yang H, Yuan J, Chan TH, Dou QP (2011) EGCG, green tea polyphenols and their synthetic analogs and prodrugs for human cancer prevention and treatment. Adv Clin Chem 53:155–177

    Article  Google Scholar 

  8. Colpitts CC, Schang LM (2014) A small molecule inhibits virion attachment to heparan sulfate- or sialic acid-containing glycans. J Virol 88:7806–7817

    Article  Google Scholar 

  9. Drut RM, Drut R (1985) Congenital herpes simplex virus infection diagnosed by cytology of aspirated tracheobronchial material. Acta Cytol 29:712–713

    Google Scholar 

  10. Gosslau A, En Jao DL, Huang MT, Ho CT, Evans D, Rawson NE, Chen KY (2011) Effects of the black tea polyphenol theaflavin-2 on apoptotic and inflammatory pathways in vitro and in vivo. Mol Nutr Food Res 55:198–208

    Article  Google Scholar 

  11. Graham BS, Snell JD, JR. (1983) Herpes simplex virus infection of the adult lower respiratory tract. Medicine (Baltimore) 62:384–393

    Article  Google Scholar 

  12. Hauber I, Hohenberg H, Holstermann B, Hunstein W, Hauber J (2009) The main green tea polyphenol epigallocatechin-3-gallate counteracts semen-mediated enhancement of HIV infection. Proc Natl Acad Sci 106:9033–9038

    Article  Google Scholar 

  13. Hedner E, Vahlne A, Hirsch JM (1990) Primary herpes simplex virus (type 1) infection delays healing of oral excisional and extraction wounds in the rat. J Oral Pathol Med 19:471–476

    Article  Google Scholar 

  14. Hegazy A, Mostafa I, Elshaier YAMM, Mahmoud SH, Abo Shama NM, Shehata M, Yahya G, Nasr NF, EL-Halawany AM, Ali MA, AliMraheilEL-ShazlyMostafa MAMAAMA (2022) Robust Antiviral Activity of Santonica Flower Extract (Artemisia cina) against Avian and Human Influenza A Viruses. In Vitro and Chemoinformatic Studies. ACS Omega 7:41212–41223

    Article  Google Scholar 

  15. Ho HY, Cheng ML, Weng SF, Leu YL, Chiu DT (2009) Antiviral effect of epigallocatechin gallate on enterovirus 71. J Agric Food Chem 57:6140–6147

    Article  Google Scholar 

  16. Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, NitscheMüllerDrostenPöhlmann WUNHAMACS (2020) SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 181:271-280.e8

    Article  Google Scholar 

  17. Jang M, Park Y-I, Cha Y-E, Park R, Namkoong S, Lee JI, Park J (2020) Tea Polyphenols EGCG and Theaflavin Inhibit the Activity of SARS-CoV-2 3CL-Protease <i>In Vitro</i>. Evidence-Based Complementary and Alternative Medicine 2020:5630838

    Article  Google Scholar 

  18. Janiuk K, Jabłońska E, Garley M (2021) Significance of NETs Formation in COVID-19. Cells. 10:151

    Article  Google Scholar 

  19. Köseler A, Sabirli R, Gören T, Türkçüer I, Kurt Ö (2020) Endoplasmic Reticulum Stress Markers in SARS-COV-2 Infection and Pneumonia: Case-Control Study. In Vivo 34:1645–1650

    Article  Google Scholar 

  20. Kumar P, Osahon O, Vides DB, Hanania N, Minard CG, Sekhar RV (2021) Severe Glutathione Deficiency, Oxidative Stress and Oxidant Damage in Adults Hospitalized with COVID-19: Implications for GlyNAC (Glycine and N-Acetylcysteine) Supplementation. Antioxidants (Basel). 11:50

    Article  Google Scholar 

  21. Kutkat O, Moatasim Y, AL-Karmalawy AA, AbulkhairGomaaEL-TaweelAbo ShamaGaballahMahmoudKayaliAliKandeilMostafa HSMRANNMMDBGMAAA (2022) Robust antiviral activity of commonly prescribed antidepressants against emerging coronaviruses: in vitro and in silico drug repurposing studies. Sci Rep 12:12920

    Article  Google Scholar 

  22. Kutok JL, Wang F (2006) Spectrum of Epstein-Barr virus-associated diseases. Annu Rev Pathol 1:375–404

    Article  Google Scholar 

  23. Kuzuhara T, Iwai Y, Takahashi H, Hatakeyama D, Echigo N (2009) Green tea catechins inhibit the endonuclease activity of influenza A virus RNA polymerase. PLoS Curr. 1:Rrn1052

    Article  Google Scholar 

  24. Laforge M, Elbim C, Frère C, Hémadi M, Massaad C, Nuss P, Benoliel J-J, Becker C (2020) Tissue damage from neutrophil-induced oxidative stress in COVID-19. Nat Rev Immunol 20:515–516

    Article  Google Scholar 

  25. Leblanc EV, Colpitts CC (2022) The green tea catechin EGCG provides proof-of-concept for a pan-coronavirus attachment inhibitor. Sci Rep 12:12899

    Article  Google Scholar 

  26. Li S, Hattori T, Kodama EN (2011) Epigallocatechin gallate inhibits the HIV reverse transcription step. Antivir Chem Chemother 21:239–243

    Article  Google Scholar 

  27. Li Z, Feng C, Dong H, Jin W, Zhang W, Zhan J, Wang S (2022) Health promoting activities and corresponding mechanism of (–)-epicatechin-3-gallate. Food Sci Human Wellness 11:568–578

    Article  Google Scholar 

  28. Liu J, Bodnar BH, Meng F, Khan AI, Wang X, Saribas S, Wang T, Lohani SC, Wang P, Wei Z, Luo J, Zhou L, Wu J, Luo G, Li Q, Hu W, Ho W (2021) Epigallocatechin gallate from green tea effectively blocks infection of SARS-CoV-2 and new variants by inhibiting spike binding to ACE2 receptor. Cell Biosci 11:168

    Article  Google Scholar 

  29. Liu Y, Du X, Chen J, Jin Y, Peng L, Wang HHX, Luo M, Chen L, Zhao Y (2020) Neutrophil-to-lymphocyte ratio as an independent risk factor for mortality in hospitalized patients with COVID-19. J Infect 81:e6–e12

    Google Scholar 

  30. Mason RJ (2020) Pathogenesis of COVID-19 from a cell biology perspective. Eur Respir J 55:2000607

    Article  Google Scholar 

  31. Menegazzi M, Campagnari R, Bertoldi M, Crupi R, Di Paola R, Cuzzocrea S (2020) Protective Effect of Epigallocatechin-3-Gallate (EGCG) in Diseases with Uncontrolled Immune Activation: Could Such a Scenario Be Helpful to Counteract COVID-19? Int J Mol Sci 21(14):5171

    Article  Google Scholar 

  32. Mettenleiter TC, Klupp BG, Granzow H (2009) Herpesvirus assembly: an update. Virus Res 143:222–234

    Article  Google Scholar 

  33. Mokra D, Adamcakova J, Mokry J (2022) Green Tea Polyphenol (-)-Epigallocatechin-3-Gallate (EGCG): A Time for a New Player in the Treatment of Respiratory Diseases? Antioxidants (Basel) 11:1566

    Article  Google Scholar 

  34. Morais Da Silva M, Lira De Lucena AS, Paiva Júnior SSL, Florêncio De Carvalho VM, Santana De Oliveira PS, Da Rosa MM, De Melo B, Rego MJ, Pitta M, Pereira MC (2022) Cell death mechanisms involved in cell injury caused by SARS-CoV-2. Rev Med Virol 32:e2292

    Article  Google Scholar 

  35. Nagle DG, Ferreira D, Zhou YD (2006) Epigallocatechin-3-gallate (EGCG): chemical and biomedical perspectives. Phytochemistry 67:1849–1855

    Article  Google Scholar 

  36. Ohishi T, Hishiki T, Baig MS, Rajpoot S, Saqib U, Takasaki T, Hara Y (2022) Epigallocatechin gallate (EGCG) attenuates severe acute respiratory coronavirus disease 2 (SARS-CoV-2) infection by blocking the interaction of SARS-CoV-2 spike protein receptor-binding domain to human angiotensin-converting enzyme 2. PLoS ONE 17:e0271112

    Article  Google Scholar 

  37. Pae M, Wu D (2013) Immunomodulating effects of epigallocatechin-3-gallate from green tea: mechanisms and applications. Food Funct 4:1287–1303

    Article  Google Scholar 

  38. Patel AR, Romanelli P, Roberts B, Kirsner RS (2007) Treatment of herpes simplex virus infection: rationale for occlusion. Adv Skin Wound Care 20:408–412

    Article  Google Scholar 

  39. Proal AD, Vanelzakker MB (2021) Long COVID or Post-acute Sequelae of COVID-19 (PASC): An Overview of Biological Factors That May Contribute to Persistent Symptoms. Front Microbiol 12:698169

    Article  Google Scholar 

  40. Queen D, Harding K (2022) COVID-19 consequences in the management of persons with wounds. Int Wound J 19:237–238

    Article  Google Scholar 

  41. Ragab SS, Sweed AMK, Elrashedy AA, Allayeh AK (2022) Design, Synthesis, Antiviral Evaluation, and Molecular Dynamics Simulation Studies of New Spirocyclic Thiopyrimidinones as Anti HCoV-229E. Chem Biodivers 19:e202200632

    Article  Google Scholar 

  42. Ronaldo Luis T (2020) Introductory Chapter: Human Herpesvirus - A Short Introduction. In: Ronaldo Luis T (ed) Human Herpesvirus Infection. Rijeka, IntechOpen

    Google Scholar 

  43. Roshdy WH, Rashed HA, Kandeil A, Mostafa A, Moatasim Y, Kutkat O, Abo Shama NM, Gomaa MR, El-Sayed IH, Guindy EL, N. M., Naguib, A., Kayali, G. & Ali, M. A. (2020) EGYVIR: An immunomodulatory herbal extract with potent antiviral activity against SARS-CoV-2. PLoS ONE 15:e0241739

    Article  Google Scholar 

  44. Seliem IA, Panda SS, Girgis AS, Moatasim Y, Kandeil A, Mostafa A, Ali MA, Nossier ES, Rasslan F, Srour AM, Sakhuja R, Ibrahim TS, Abdel-Samii ZKM, Al-Mahmoudy AMM (2021) New quinoline-triazole conjugates: Synthesis, and antiviral properties against SARS-CoV-2. Bioorg Chem 114:105117

    Article  Google Scholar 

  45. Singh BN, Shankar S, Srivastava RK (2011) Green tea catechin, epigallocatechin-3-gallate (EGCG): mechanisms, perspectives and clinical applications. Biochem Pharmacol 82:1807–1821

    Article  Google Scholar 

  46. Sriram N, Kalayarasan S, Sudhandiran G (2008) Enhancement of Antioxidant Defense System by Epigallocatechin-3-gallate during Bleomycin Induced Experimental Pulmonary Fibrosis. Biol Pharm Bull 31:1306–1311

    Article  Google Scholar 

  47. Sriram N, Kalayarasan S, Sudhandiran G (2009) Epigallocatechin-3-gallate augments antioxidant activities and inhibits inflammation during bleomycin-induced experimental pulmonary fibrosis through Nrf2–Keap1 signaling. Pulm Pharmacol Ther 22:221–236

    Article  Google Scholar 

  48. Sriram N, Kalayarasan S, Sudhandiran G (2009) Epigallocatechin-3-gallate exhibits anti-fibrotic effect by attenuating bleomycin-induced glycoconjugates, lysosomal hydrolases and ultrastructural changes in rat model pulmonary fibrosis. Chem Biol Interact 180:271–280

    Article  Google Scholar 

  49. Steinmann J, Buer J, Pietschmann T, Steinmann E (2013) Anti-infective properties of epigallocatechin-3-gallate (EGCG), a component of green tea. Br J Pharmacol 168:1059–1073

    Article  Google Scholar 

  50. Tsai MJ, Chang WA, Liao SH, Chang KF, Sheu CC, Kuo PL (2019) The Effects of Epigallocatechin Gallate (EGCG) on Pulmonary Fibroblasts of Idiopathic Pulmonary Fibrosis (IPF)-A Next-Generation Sequencing and Bioinformatic Approach. Int J Mol Sci 20:1958

    Article  Google Scholar 

  51. Vázquez-Calvo Á, Jiménez De Oya N, Martín-Acebes MA, Garcia-Moruno E, Saiz JC (2017) Antiviral Properties of the Natural Polyphenols Delphinidin and Epigallocatechin Gallate against the Flaviviruses West Nile Virus, Zika Virus, and Dengue Virus. Front Microbiol 8:1314

    Article  Google Scholar 

  52. Von Knethen A, Heinicke U, Laux V, Parnham MJ, Steinbicker AU, Zacharowski K (2022) Antioxidants as Therapeutic Agents in Acute Respiratory Distress Syndrome (ARDS) Treatment-From Mice to Men. Biomedicines 10:98

    Article  Google Scholar 

  53. Wang Y, Shao S, Xu P, Yang X, Qian L (2011) Catechin-enriched green tea extract as a safe and effective agent for antimicrobial and anti-inflammatory treatment. Afr J Pharm Pharmacol 5:1452–1461

    Article  Google Scholar 

  54. Wang YQ, Li QS, Zheng XQ, Lu JL, Liang YR (2021) Antiviral Effects of Green Tea EGCG and Its Potential Application against COVID-19. Molecules 26:3962

    Article  Google Scholar 

  55. Weber C, Sliva K, Von Rhein C, Kümmerer BM, Schnierle BS (2015) The green tea catechin, epigallocatechin gallate inhibits chikungunya virus infection. Antiviral Res 113:1–3

    Article  Google Scholar 

  56. Xu FW, Lv YL, Zhong YF, Xue YN, Wang Y, Zhang LY, Hu X, Tan WQ (2021) Beneficial Effects of Green Tea EGCG on Skin Wound Healing: A Comprehensive Review. Molecules 26:6123

    Article  Google Scholar 

  57. Xu J, Gu W, Li C, Li X, Xing G, Li Y, Song Y, Zheng W (2016) Epigallocatechin gallate inhibits hepatitis B virus via farnesoid X receptor alpha. J Nat Med 70:584–591

    Article  Google Scholar 

  58. Xu J, Xu Z, Zheng W (2017) A Review of the Antiviral Role of Green Tea Catechins. Molecules 22:1337

    Article  Google Scholar 

  59. Zawani M, Fauzi MB (2021) Epigallocatechin Gallate: The Emerging Wound Healing Potential of Multifunctional Biomaterials for Future Precision Medicine Treatment Strategies. Polymers (Basel) 13:3656

    Article  Google Scholar 

Download references

Acknowledgements

We acknowledge the funding from the Deanship of Scientific Research at Jouf University under Grant Number (DSR2022-RG-0152).

Funding

This work was funded by the Deanship of Scientific Research at Jouf University under Grant Number (DSR2022-RG-0152).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, A.M., G.M-H., A.M.A. and A.K.A.; methodology, A.M., G.M-H., H.A.E., E.A.M., A.Y.E., S.H.A., A.M.A., and A.K.A.; formal analysis, G.M-H., A.M., H.A.E., A.Y.E., S.H.A., A.M.A. and A.K.A.; investigation, G.M-H., A.M., A.M.A. and A.K.A.; data curation, G.M-H., A.M., and A.K.A.; Supervision, G.M-H., A.M., A.M.A. and A.K.A.; writing—original draft preparation, A.M., A.M.A. and A.K.A.; writing—review and ed-iting, G.M-H., A.M., A.M.A. and A.K.A. All authors have read and agreed to the published version of the manuscript.

Corresponding authors

Correspondence to Ahmed Mostafa or Gomaa Mostafa-Hedeab.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the review.

Additional information

Publisher’s Note

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

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

Mostafa, A., Mostafa-Hedeab, G., Elhady, H.A. et al. Dual action of epigallocatechin-3-gallate in virus-induced cell Injury. J Genet Eng Biotechnol 21, 145 (2023). https://doi.org/10.1186/s43141-023-00624-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43141-023-00624-4

Keywords