Next Article in Journal
Complete Genome Sequences of Four Mycobacteriophages Involved in Directed Evolution against Undisputed Mycobacterium abscessus Clinical Strains
Previous Article in Journal
Identifying Active Rather than Total Methanotrophs Inhabiting Surface Soil Is Essential for the Microbial Prospection of Gas Reservoirs
Previous Article in Special Issue
Enhanced Anti-Herpetic Activity of Valacyclovir Loaded in Sulfobutyl-ether-β-cyclodextrin-decorated Chitosan Nanodroplets
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Antiherpetic Activity of a Root Exudate from Solanum lycopersicum

1
Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
2
Center for Translational Research on Autoimmune and Allergic Disease (CAAD), 28100 Novara, Italy
3
Department of Chemistry, University of Turin, 10125 Turin, Italy
4
Department of Agricultural, Forestry, and Food Sciences, University of Turin, 10095 Turin, Italy
5
Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Microorganisms 2024, 12(2), 373; https://doi.org/10.3390/microorganisms12020373
Submission received: 29 December 2023 / Revised: 5 February 2024 / Accepted: 9 February 2024 / Published: 11 February 2024

Abstract

:
The rise of drug resistance to antivirals poses a significant global concern for public health; therefore, there is a pressing need to identify novel compounds that can effectively counteract strains resistant to current antiviral treatments. In light of this, researchers have been exploring new approaches, including the investigation of natural compounds as alternative sources for developing potent antiviral therapies. Thus, this work aimed to evaluate the antiviral properties of the organic-soluble fraction of a root exudate derived from the tomato plant Solanum lycopersicum in the context of herpesvirus infections. Our findings demonstrated that a root exudate from Solanum lycopersicum exhibits remarkable efficacy against prominent members of the family Herpesviridae, specifically herpes simplex virus type 1 (HSV-1) (EC50 25.57 µg/mL, SI > 15.64) and human cytomegalovirus (HCMV) (EC50 9.17 µg/mL, SI 32.28) by inhibiting a molecular event during the herpesvirus replication phase. Moreover, the phytochemical fingerprint of the Solanum lycopersicum root exudate was characterized through mass spectrometry. Overall, these data have unveiled a novel natural product with antiherpetic activity, presenting a promising and valuable alternative to existing drugs.

Graphical Abstract

1. Introduction

Herpesviruses pose a significant threat to human health, causing chronic and recurring infections that profoundly impact the quality of life for affected individuals. Notable members of this viral family include herpes simplex virus type 1 (HSV-1) and human cytomegalovirus (HCMV) [1,2]. Many members of the Herpesviridae family exhibit high seroprevalence globally; HCMV reaches more than 50% of the general population, and HSV, more than 60%. Around 500 million people worldwide, aged 15 to 50, have genital infections caused by HSV-1 or HSV-2, while the incidence of cold sores is second globally only to the common cold. HCMV infection stands as the prevailing congenital infection globally, with an estimated occurrence in developed nations ranging from 0.6% to 0.7% of all live births. This translates to around 60,000 neonates born annually with HCMV infection in the Western world. Notably, in developing countries, the prevalence is even more pronounced, affecting between 1% and 5% of all live births [2,3,4]. The efficacy of existing treatments is compromised by resistant strains of herpesviruses, highlighting the pressing need to explore alternative compounds and formulations [5,6,7]. Therefore, the quest for new antiherpetic drugs is a critical global challenge not only to alleviate the suffering caused by herpesvirus, but also to counter drug-resistant herpesvirus strains, improve treatment outcomes, and tackle the evolving challenges exacerbated by the emergence of viral escape mutants.
Within this framework, herbal medicines and purified natural products derived from plants, marine organisms, and other natural sources have garnered considerable attention as a rich resource for the development of innovative antiviral drugs. This includes a diverse array of substances, including essential oils, plant extracts, small peptides from animal sources, bacteriocins, and several categories of plant compounds (such as alkaloids, triterpenoids, flavonoids, and phenols) renowned for their antimicrobial and antiviral properties [8,9,10]. The use of natural compounds as antivirals presents several advantages over synthetic drugs. Indeed, they are often derived from renewable sources, making them more sustainable and environmentally friendly. Moreover, these compounds may have fewer adverse side effects compared to synthetic drugs, as they have evolved alongside living organisms and are more compatible with human biology [11,12].
Solanum lycopersicum, commonly referred to as the tomato plant, is a species of flowering plant that belongs to the Solanaceae family. It ranks as the second most widely cultivated and consumed vegetable worldwide, making it both an essential model organism for plant research and a significant commercial crop. In addition to its noteworthy nutritional value, Solanum lycopersicum has been studied for its potential medicinal properties which have been recognized and utilized for centuries [13,14,15,16]. Notably, tomato extracts contain a diverse array of bioactive compounds, such as flavonoids and carotenoids. These specialized metabolites accumulate in various tissues of the tomato plant and are released as root exudates and volatiles [17]. Tomato root exudates are rich in organic acids, sugars, and amino acids. Among these, glucose and fructose stand out as the primary sugars; malic, citric, and succinic acids are the predominant organic acids [18]; and glutamic acid, aspartic acid, leucine, isoleucine, and lysine are the main amino acids [19]. The secretion levels differ based on the growth phase of the tomato and the presence of particular microorganisms [20].
These compounds impart a significant nutraceutical value to tomatoes, positioning them as functional foods that can positively impact various pathological conditions [16]. One notable example is their ability to reduce the production of pro-inflammatory molecules, thereby mitigating inflammation and potentially avoiding the onset of chronic inflammatory diseases [21]. Furthermore, tomatoes have also garnered attention for their potential cardiovascular benefits. The abundant potassium content found in Solanum lycopersicum assists in regulating blood pressure by counteracting the effects of sodium, thus supporting a healthy cardiovascular system. Lastly, in terms of medicinal properties, it is suggested that it may contribute to a reduction in cancer risks, particularly for lung and prostate cancers [22,23].
Despite numerous investigations into the therapeutic properties of tomatoes, there is a lack of studies exploring their potential antiviral effects, particularly those of root exudates. Interestingly, the antiviral efficacy of tomatidine, a steroidal alkaloid obtained from the stem and leaves of unripe, green tomatoes, has been demonstrated against all four dengue virus (DENV) serotypes and Zika virus (ZIKV) in the Huh7 in vitro model. It disrupts various stages of DENV replication, especially post-virus–cell binding and internalization. The cellular activating transcription factor 4 (ATF4) may enhance the antiviral effect, yet it is not entirely responsible for it. Notably, no antiviral activity was detected for West Nile virus (WNV), another mosquito-borne flavivirus [24]. Tomatidine and its structural derivatives solasodine and sarsasapogenin were also effective against multiple chikungunya virus (CHIKV) strains, primarily during the early stages of infection [25]. In the context of DNA viruses, tomatine displayed inhibitory activity against HSV-1, possibly through the interaction of the sugar moiety of the glycosidic chain with the viral envelope [26].
In this study, recognizing the need to continue searching for novel antiviral agents, we aimed to investigate the in vitro antiviral activity of a root exudate derived from Solanum lycopersicum against two members of the herpesvirus family (specifically HSV-1 and HCMV). Additionally, the phytochemical fingerprint of the exudate was determined by ESI-MS analyses, disclosing the occurrence of several bioactive compounds (carotenoids, phytosterols, and polyphenols) which might play a synergic role in determining the antiviral activity of the tomato root exudate. In summary, these data highlight Solanum lycopersicum as a natural product with the potential to serve as a valuable alternative to existing antiviral drugs.

2. Materials and Methods

2.1. Plant Material and Tomato Extract Preparation

Tomato (Solanum lycopersicum cv. M82) seeds were surfaced-sterilized in 4% sodium hypochlorite containing 0.02% (v/v) Tween 20, washed thoroughly with sterile water, and germinated for 2 days in a plate on moistened filter paper at 25 °C in the darkness. Subsequently, 4-week-old plants were transferred in an aeroponic system and grown with Hogland nutrient solution (0.2 μM ZnSO4, 0.2 μM CuSO4, 1 mM Ca(NO3)2, 80 μM KH2PO4, 250 μM KNO3, 20 μM FeNa EDTA, 1.8 μM MnCl2, 0.2 μM Co(NO3)2, 9 μM H3BO3, 0.2 μM NiSO4, 1 mM MgSO4, pH 6.0). The plants were cultured in a growth chamber set at the following parameters: 16/8 h day/night cycle, 25 °C, 65% humidity, and 200 µmol s–1 m–2 of photosynthetic photon flux density (PPFD). The nutrient solution was renewed and alternated with a version containing an equimolar quantity of KCl instead of KH2PO4. The nutrient solution containing the root exudates was collected and filtered twice a week by a 0.2 mm sieve. Subsequently, the root exudate was loaded onto the preequilibrated column (Grace Pure C18-Fast 500 mg/3 mL SPE) and eluted with 5 mL of 1:1 water/acetone solution. Finally, the eluted solution was dried with a rotary evaporator and stocked at −20 °C (Figure 1) [27]. At the time of use, 10 mg of lyophilized root exudate was dissolved in 1 mL of DMSO/H2O mixture (1:1) and subsequently diluted in the culture medium.

2.2. Phytochemical Analysis

Acetonitrile (HPLC-grade) and formic acid (mass-spectrometry-grade, 98%) were purchased from Sigma Aldrich. The tomato root exudate sample was diluted in acetonitrile (1.0 mg/mL) with 0.1% formic acid as a charge carrier.
Direct-infusion positive ESI-MS analysis was performed on a 3200 QTRAP® LC-MS/MS System (AB Sciex) equipped with a syringe pump. The sample was infused at a flow rate of 10 µL/min. The paramete, rs adopted for the ESI source were as follows: source voltage 5.0 kV; heated capillary temperature 350 °C; N2 curtain gas flow rate 20 (arbitrary units); declustering potential 20 V; entrance potential 10 V. Total ion current (TIC) mode was used to record the abundances of the ionized adducts. In the full-scan mode, masses were scanned as centroid data from m/z 100 to 700 at a rate of 1 scan/s (averaged over 5 min). Data acquisition and processing and instrumental control were performed using the Analyst®® software package version 1.7 (AB Sciex).

2.3. Cells and Viruses

African green monkey kidney cells (VERO, ATCC CCL-81™) and primary human foreskin fibroblasts (HFFs, ATCC SCRC-1041™) were propagated in Dulbecco’s Modified Eagle Medium (DMEM; Sigma-Aldrich, Milan, Italy) complemented with 1% streptomycin/penicillin solution (Sigma-Aldrich) and 10% fetal bovine serum (heat-inactivated) (Sigma-Aldrich).
The clinical isolate of HSV-1 (obtained from Valeria Ghisetti, “Amedeo di Savoia” Hospital, Turin, Italy) and the AD169 HCMV strain (ATCC VR538) were grown and titrated by a standard plaque assay on VERO and HFF cells, respectively, on a 96-well plate (~30,000 cells/well), as previously described [28,29].

2.4. Cytotoxicity Assay

VERO and HFF cells were seeded in a 96-well culture plate (~30,000 cells/well) and exposed to increasing concentrations of either tomato extract or vehicle control (DMSO/H2O mixture 1:1). After 48 h (for VERO) or 144 h (for HFF) of incubation, cell viability was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) (Sigma-Aldrich) assay, as previously described [30].

2.5. Virus Yield Reduction Assay

VERO and HFF cells were plated in 24-well plates (~200,000 cells/well) and pre-treated with different concentrations of tomato extracts, vehicle, or the reference drugs Acyclovir (ACV, Sigma-Aldrich) for HSV-1 and Ganciclovir (GCV, Sigma-Aldrich) for HCMV for 1 h at 37 °C. They were then infected with HSV-1 (VERO) or HCMV (HFF) at an MOI of 0.1 plaque-forming units per cell (PFU/cell) in the presence of treatments. Following virus adsorption (2 h at 37 °C), the viral inoculum was removed, and the cultures were maintained in a medium that contained the extract for 48 h (HSV-1) or 144 h (HCMV). Cells and supernatants (combined) were then collected and lysed using three freeze (liquid nitrogen)/thaw (37 °C) cycles. Virus replication was assessed by titrating the infectivity of the samples obtained from the virus yield reduction assay with a standard plaque assay on VERO (HSV-1) and HFF (HCMV) previously plated in a 96-well plate (~30,000 cells/well). After 48 h (HSV-1) or 144 h (HCMV), cells were fixed and stained with a crystal violet solution (0.1% crystal violet, 20% ethanol). Plaques were microscopically counted, and the mean plaque counts for each drug concentration were expressed as PFU/mL. The concentration that produced a 50% reduction in plaque formation (EC50) was determined for each test by nonlinear regression (curve fitting analysis) in GraphPad Prism software version 8.02 for Windows. The selectivity index (SI) reflects the CC50-to-EC50 ratio.

2.6. Time of Addition Assay

VERO and HFF cells were seeded in 24-well plates (~200,000 cells/well) and treated with various concentrations of tomato extract or vehicle solution at different time points: (i) 1 h pre-infection as pre-treatment, (ii) only during the 2 h of viral adsorption, (iii) post-infection until sample collection. In all conditions, cells were infected with HSV-1 or HCMV (MOI 0.1 PFU/cell). After 48 h (VERO) or 144 h (HFF), cells and supernatants were collected and disrupted using three freeze (liquid nitrogen)/thaw (37 °C) cycles. The level of virus replication was then assessed by titrating the infectivity of the samples by a plaque assay as described in the virus yield reduction assay section.

2.7. Western Blot Analysis

VERO and HFF cells were seeded in 6-well plates (~106 cells/well), treated as described in the virus yield reduction assay section, and infected at an MOI of 1. Cells were collected at different time points, and whole-cell protein extracts were prepared using Radioimmunoprecipitation Assay (RIPA) buffer. Then, 30 µg of protein extracts were subjected to electrophoresis on sodium dodecyl sulfate–polyacrylamide (SDS) gels and transferred to Immobilon-P membranes (Biorad). Membranes were blocked with 5% nonfat dry milk in TBS-Tween 0.05% and incubated overnight at 4 °C with the primary antibodies. The following mouse monoclonal primary antibodies were used: anti-HSV-1 ICP4 (clone 10F1, H1A021-100, Virusys Corporation, Taneytown, MD, USA), anti-HSV-1/2 gD (clone 2C10, HA025-100, Virusys Corporation), anti-HCMV IEA (clone CH160, P1215, Virusys Corporation), pp65 (clone 3A12, CA003-100, Virusys Corporation), pp28 (clone 5C3, CA004-100, Virusys Corporation), anti-Actin (clone C4, MAB1501, Sigma-Aldrich) (all at 1:1000 dilution in 5% nonfat dry milk, TBS-Tween 0.05%). After washing with TBST buffer (500 mM NaCl, 20 mM Tris pH 7.4, 0.05% Tween 20), the membranes were incubated with an HRP-conjugated anti-mouse secondary antibody (1:3000 diluted, Sigma-Aldrich) for 2 h at room temperature, and immunocomplexes were visualized using an enhanced chemiluminescence detection kit (SuperSignal West Pico Chemiluminescent Substrate, Thermo SCIENTIFIC, Waltham, MA, USA).

2.8. DNA Extraction and Viral Load

VERO and HFF cells were seeded in 24-well plates (~200,000 cells/well), treated as described in the virus yield reduction assay section, and infected at an MOI of 1. After 24 (HSV-1) or 72 (HCMV) hours, cells were collected, and the cellular-associated DNA was isolated using the TRI Reagent solution (Sigma-Aldrich) according to the manufacturer’s instructions. The extracted viral DNA was quantified by quantitative real-time PCR (qPCR) analysis using a CFX Touch Real-Time PCR Detection System (BioRad, Hercules, CA, US). The viral DNA copy numbers were quantified using primers to amplify a segment of the IE1 gene for HCMV (Fw, 5′-TCAGTGCTCCCCTGATGAGA-3′; Rv, 5′-GATCAATGTGCGTGAGCACC-3′) or gE for HSV (Fw, 5′-TGTCTGTATCAGCCGCAGC-3′; Rv, 5′-TTCTGGAACACCCCGCGTA-3′). Intracellular viral DNA copy numbers were normalized to GAPDH (Fw, 5′-AGTGGGTGTCGCTGTTGAAGT-3′; Rv, 5′-AACGTGTCAGTGGTGGACCTG-3′). To create a standard curve for each analysis, genomic DNA mixed with a gE2- or IE1-encoding plasmid (pAcUW51-CgE, a gift from Pamela Bjorkman; Addgene plasmid #13762, http://n2t.net/addgene:13762 accessed on 31 December 2023, RRID: Addgene_13762 [31] (Addgene, Watertown, MA, USA; pSG-IE72, available at the Department of Public Health and Pediatric Sciences, Turin, Italy) was serially diluted and analyzed in parallel.

2.9. Statistical Analysis

The statistical tests were performed using GraphPad Prism version 8.0.2 for Windows (GraphPad, Boston, MA, USA. Data were presented as the mean value and standard error of the mean (SEM). Differences were considered statistically significant for p < 0.05 (p < 0.05 *; p < 0.01 **; p < 0.001 ***; p < 0.0001 ****).

3. Results and Discussion

In this work, we aimed to study the antiherpetic activity of a root exudate of Solanum lycopersicum in vitro (named tomato extract). Initially, we focused on HSV-1, an alpha-herpesvirus highly transmissible and widespread in the population [32,33]. First, the organic extract was prepared from the root exudates of the plants, as described in the materials and methods section. Subsequently, its impact on cellular viability was assessed using a standard MTT assay on uninfected VERO cells, treated with increasing extract concentrations (10–400 µg/mL) or an equal volume of control vehicle (CTRL, DMSO/H2O mixture 1:1). The results, depicted in Figure 2A, revealed that the half-maximal cytotoxic concentration (CC50) after 48 h of treatment exceeded 400 µg/mL, indicating a lack of significant cytotoxicity at the tested concentrations. To evaluate the potential antiviral activity of the tomato root exudate, we first performed a virus yield reduction assay by treating VERO cells in the presence of serial dilutions of the exudate before, during, and after viral infection to generate a dose–response curve. The extent of HSV-1 replication was then assessed by titrating the infectivity of both supernatants and cell-associated viruses combined through a standard plaque assay in VERO cells. The results presented in Figure 2B demonstrated that the exudate was endowed with anti-HSV-1 activity with the half-maximal inhibitory concentration (EC50) value of 25.57 (±8.51) µg/mL. Based on the above results, the SI for HSV-1 is >15.64. Noteworthily, at the highest non-toxic concentration of tomato extract (400 µg/mL), the infectious virus titer was reduced to ~105 PFU/mL, which corresponds to a reduction of over 99% when compared to the vehicle control. Therefore, this concentration was employed in the subsequent experiments. The widely employed antiviral drug ACV was included as a positive control (44.4 µM [34]). As expected, its presence resulted in a complete reduction in viral replication (Figure 2B). To further confirm the antiviral activity, we examined the impact of the treatment on viral protein expression by Western blot analysis. Immunoblotting with specific antibodies was performed to analyze the expression patterns of viral proteins, particularly immediate early/early (ICP4) and late (gD) viral products. A noticeable decrease in viral protein levels was observed at each time point in cells treated with tomato exudate or ACV (Figure 2C). These results suggest that the root exudate interferes with a molecular event that occurs in the early stages of the HSV-1 replication cycle. To further analyze the impact of the tomato exudate on viral DNA replication and the production of new viral genomes, we performed a quantitative real-time PCR (qPCR) analysis. As depicted in Figure 2D, a significant reduction in intracellular copies of the HSV-1 genome was observed after 24 h of treatment, providing additional evidence for the inhibitory impact of the root exudate on viral replication.
To determine whether the antiviral activity of Solanum lycopersicum was limited to HSV-1 or encompassed other herpesviruses, we investigated its efficacy on the replication of HCMV, which belongs to the Betaherpesvirinae subfamily (Figure 3). An MTT viability assay was conducted on HFF cells after 144 h of treatment to establish the CC50 dose, which was calculated to be 296.00 (±80) µg/mL (Figure 3A). Through a virus yield reduction assay, performed with serial dilutions of root exudate, we confirmed the antiviral activity of the compound also against HCMV, with an EC50 value of 9.17 (±0.65) µg/mL (Figure 3B). The SI for HCMV is 32.28. GCV (25 µM [35]) was included as a positive control.
Similarly to the approach taken for HSV-1, cells underwent Western blot and qPCR analysis after exposure to the extract at concentrations of 100 µg/mL (highly effective non-toxic concentrations, over 99% inhibition). Western blot analysis revealed a decrease in viral protein expression, including immediate early (IEA), early–late (pp65), and late (pp28) proteins, suggesting an overall inhibitory effect on HCMV replication (Figure 3C). Finally, we assessed the extent of intracellular viral genome replication upon treatment with the extract by a qPCR analysis at 72 hpi. Consistently, the results shown in Figure 3D highlight a significant reduction in the viral genome copy number compared to vehicle-treated samples.
Next, to identify which phase of the viral replication cycle was mainly affected by the treatment, serial dilutions of extract were added to VERO or HFF cells at different time schedules: (i) one hour before infection (pre-adsorption stage), (ii) for 2 h during viral adsorption and then removed (adsorption stage), (iii) after the removal of viral inoculum (post-adsorption stage). For each condition, the infected samples were collected at 48 hpi for HSV-1 and 144 hpi for HCMV. Plaque assay titration revealed that the most substantial antiviral effect occurred when the root extract was added after the infection (Figure 4C), compared to the pre-adsorption (Figure 4A) or the adsorption (Figure 4B) conditions.
Our results align with prior research that underscores the use of various Solanum-genus plants, including Solanum dulcamara, Solanum lyratum, and Solanum nigrum, as traditional anti-herpes agents since ancient times [36]. Importantly, our study extends this activity for the first time to HCMV, representing another member of the Herpesviridae family. Studies indicate that Solanum steroidal glycosides, particularly spirostanol glycosides, inhibit HSV-1. It has been suggested that the activity is influenced by the type of oligosaccharide moiety, but the underlying mechanism remains unclear [36]. Additionally, extracts from S. paniculatum leaves demonstrated efficacy against HSV-1 in a VERO cell in vitro model, albeit with a lower SI compared to our product. In this context, the mechanism of action has not been elucidated [37]. Notably, Solanaceae glycoalkaloids insert glycones into the viral envelope, causing HSV-1 virion leakage [26]. While we cannot completely rule out a similar mechanism for root exudate extracts, our observations suggest only minimal inhibition when the compound is added during viral pre-adsorption and adsorption (i.e., attachment and entry phases). Since the primary inhibitory effect of the extract was prominently observed when added post-infection, we can infer that the root extract interferes with a molecular event during the herpesvirus replication stage. These findings are consistent with viral protein expression and qPCR results (Figure 2 and Figure 3, panels C and D) and align with earlier studies on DENV [24] and CHIKV [25] viral models.
Finally, the phytochemical composition of the Solanum lycopersicum root exudate was investigated through mass spectrometry. Specifically, electrospray ionization (ESI-MS) was employed, enabling the acquisition of mass spectra with a low degree of fragmentation and the qualitative characterization of the complex mixture, consisting of small chemicals and biomolecular components [38]. The full-scan positive ion ESI-MS spectrum is depicted in Figure 5, with detailed information on the major components presented in Table 1.
Five main classes of biologically derived compounds were identified in the ESI-MS spectrum belonging to different substances (see Table 1) commonly found in tomato extracts: carotenoids, glycoalkaloids, phytosterols, flavonoids, and amino acids [39,40]. The first class, carotenoids, includes the typical fragment of ionized tetraterpene derivatives lycopene and β-carotene (M.W. 536.88 g/mol, m/z = 537 [M + H]+), while doubly charged adducts can be found for lutein (M.W. 568.87 g/mol, m/z = 296 [M + H + Na]2+), phytoene (M.W. 544.95 g/mol, m/z = 282 [M + H + NH4]2+), and phytofluene (M.W. 542.94 g/mol, m/z = 280 [M + H + NH4]2+). Glycoalkaloid derivatives identified were α-tomatine, with the molecular fragment at m/z = 529 which might result from the formation of a doubly charged adduct (M.W. 1034.18 g/mol, [M + H + Na]2+), its corresponding aglycone tomatidine (M.W. 415.66 g/mol, m/z = 416 [M + H]+), and lycoperoside H (M.W. 1049.54 g/mol, m/z = 525 [M + 2H]2+). The ionized adduct at m/z = 416 can be assigned to the phytosterol derivative β-sitosterol (M.W. 414.72 g/mol, [M + H]+). The fourth class of polyphenolic compounds (flavonoids) identified in the tomato root exudate includes myricetin (M.W. 318.04 g/mol, m/z = 336 [M + NH4]+ and m/z = 168 [M + H + NH4]2+), isorhamnetin (M.W. 316.26 g/mol, m/z = 334 [M + NH4]+), quercetin (M.W. 302.24 g/mol, m/z = 320 [M + NH4]+), and kaempferol (M.W. 286.23 g/mol, m/z = 287 [M + H]+). Lastly (glyco)amino acid derivatives N-acetylglucosamine-asparagine (GlcNAc-(1→N)-Asn, M.W. 335.31 g/mol, m/z = 353 [M + NH4]+), N-acetyl-L-hydroxyproline (AHYP, M.W. 173.17 g/mol, m/z = 174 [M + H]+), and homocysteine (M.W. 135.18 g/mol, m/z = 136 [M + H]+) were identified.
The results of this qualitative analysis show that the root exudate of Solanum lycopersicum contains a number of phytochemicals with recognized antiviral activity. In particular, some key identified phytochemicals in the root exudate possess antiherpetic properties, as demonstrated for lycopene (EC50 = 22.86 μg/mL for HSV-1) [41], β-sitosterol (EC50 = 2.7 μg/mL for HSV-2) [42], tomatidine [26], and several flavonoids [43,44], including quercetin (EC50 = 52.90 μg/mL for HSV-1 and EC50 = 70.01 μg/mL for HSV-2) [45,46], kaempferol [47], and myricetin [44]. Furthermore, natural antioxidants, such as bioactive carotenoids (lycopene, β-carotene, lutein, phytoene, phytofluene), phytosterols (β-sitosterol), and flavonoids (quercetin, kaempferol, myricetin,) have a beneficial role in human health and chronic diseases owing to their ability to modulate the activity of specific enzymes or inhibit the production of reactive oxygen species (ROS) acting as free radical scavengers [48,49,50]. Additionally, it is well known that the pharmacological profile of flavonoids, carotenoids, glycoalkaloids, and phytosterols includes anticancer, anti-inflammatory, anti-allergenic, antithrombotic, antimicrobial, antioxidant, vasodilator, and cardioprotective effects [51,52,53,54,55].
Since phytoextracts are mixtures of secondary metabolites, the pharmacological behavior of plant extracts is often due to the synergic effect of two or more active constituents rather than a single component. Indeed, the results presented in this work can be rationalized assuming that the biologically active compounds detected in the Solanum lycopersicum root exudate might play a synergic role in the antiviral activity against herpesviruses. The combination of the antiherpetic activity of the detected phytochemicals with their antioxidant properties and other pharmacological profiles make this extract a promising candidate for the development of the root exudate of Solanum lycopersicum as a new combined phytomedicine. Overall, these findings could potentially contribute to progress in the field of antiviral phytotherapy, which has experienced a resurgent interest in the field of antiviral drug discovery owing to the increasing urge to contest the development of drug resistance [56].

4. Conclusions

In conclusion, our research highlights the potential of tomato plants as a natural product exhibiting antiviral activity against herpesviruses. The exudate from Solanum lycopersicum root significantly inhibited HSV-1 and HCMV replication in VERO and HFF cells, with EC50 values of 25.57 and 9.17 µM, respectively. The SIs of >15.64 for HSV-1 and 32.28 for HCMV imply a substantial therapeutic window, indicating that the drug can effectively inhibit the target while maintaining a relatively low cytotoxic impact on host cells, making it a promising candidate for further preclinical and clinical development. Subsequent protein expression analysis and time-of-addition experiments revealed that the antiviral effect is mainly produced in post-infection conditions, during the viral replication phase, as no or a limited antiviral effect was detectable for the pre-infection and during-infection treatments. These findings suggest that Solanum lycopersicum could be a promising alternative to current antiviral drugs. Further investigation is necessary to understand the specific mechanisms of action, the efficacy in vivo, and the pharmacokinetics of the extract. Finally, it will be crucial to identify the more active molecules within the root exudate responsible for the observed antiviral effects. Nevertheless, this study offers valuable insights into the potential of Solanum lycopersicum as a source of natural antiviral agents in the ongoing efforts to combat viral infections and uphold public health safety.

Author Contributions

Conceptualization, M.B. (Matteo Biolatti), M.B. (Marco Blangetti), I.V. and V.D.; data curation, M.B. (Matteo Biolatti), M.B. (Marco Blangetti), G.B., D.A. and V.D.; formal analysis, F.S., I.V., V.D., M.B. (Marco Blangetti), C.A., L.T. and S.P.; funding acquisition, F.S., M.D.A., C.P., M.B. (Marco Blangetti), M.B. (Matteo Biolatti) and V.D.; investigation, M.B. (Matteo Biolatti), G.B. and D.A.; methodology, M.B. (Matteo Biolatti), M.B. (Marco Blangetti), G.B., D.A., E.C., C.A., L.T., S.P. and F.G.; writing—original draft, M.B. (Matteo Biolatti), M.B. (Marco Blangetti), V.D., D.A. and G.B.; writing—review and editing, C.P., M.D.A., F.S. and I.V. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the University of Turin (RILO2021, RILO2022, and RILO2023 to M.D.A., V.D., M.B. (Matteo Biolatti), F.S., C.P., M.B. (Marco Blangetti) and I.V.), by the Italian Ministry of University and Research—MUR (PRIN Projects 2017ALPCM and 2022S3AZCC to V.D., 2022N7EXJM to Matteo Biolatti), by the “Cassa di Risparmio” of Turin Foundation (RF = 2021.1745 to Matteo Biolatti), by EU funding within the MUR PNRR Extended Partnership initiative on Emerging Infectious Diseases (Project No. PE00000007, INF-ACT to M.D.A. and M.B. (Marco Blangetti)), and by Project CH4.0 under the MUR program “Dipartimenti di Eccellenza 2023–2027” (CUP: D13C22003520001, to C.P., M.B. (Marco Blangetti) and D.A.).

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflicts 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 results.

References

  1. Zhu, S.; Viejo-Borbolla, A. Pathogenesis and Virulence of Herpes Simplex Virus. Virulence 2021, 12, 2670–2702. [Google Scholar] [CrossRef]
  2. Gugliesi, F.; Coscia, A.; Griffante, G.; Galitska, G.; Pasquero, S.; Albano, C.; Biolatti, M. Where Do We Stand after Decades of Studying Human Cytomegalovirus? Microorganisms 2020, 8, E685. [Google Scholar] [CrossRef]
  3. Chayavichitsilp, P.; Buckwalter, J.V.; Krakowski, A.C.; Friedlander, S.F. Herpes Simplex. Pediatr. Rev. 2009, 30, 119–129, quiz 130. [Google Scholar] [CrossRef] [PubMed]
  4. James, S.H.; Kimberlin, D.W. Neonatal Herpes Simplex Virus Infection: Epidemiology and Treatment. Clin. Perinatol. 2015, 42, 47–59, viii. [Google Scholar] [CrossRef]
  5. Walti, C.S.; Khanna, N.; Avery, R.K.; Helanterä, I. New Treatment Options for Refractory/Resistant CMV Infection. Transpl. Int. 2023, 36, 11785. [Google Scholar] [CrossRef] [PubMed]
  6. Chen, S.-J.; Wang, S.-C.; Chen, Y.-C. Antiviral Agents as Therapeutic Strategies Against Cytomegalovirus Infections. Viruses 2019, 12, 21. [Google Scholar] [CrossRef] [PubMed]
  7. Piperi, E.; Papadopoulou, E.; Georgaki, M.; Dovrat, S.; Bar Illan, M.; Nikitakis, N.G.; Yarom, N. Management of Oral Herpes Simplex Virus Infections: The Problem of Resistance. A Narrative Review. Oral Dis. 2023. [Google Scholar] [CrossRef] [PubMed]
  8. Boswell, Z.; Verga, J.U.; Mackle, J.; Guerrero-Vazquez, K.; Thomas, O.P.; Cray, J.; Wolf, B.J.; Choo, Y.-M.; Croot, P.; Hamann, M.T.; et al. In-Silico Approaches for the Screening and Discovery of Broad-Spectrum Marine Natural Product Antiviral Agents against Coronaviruses. IDR 2023, 16, 2321–2338. [Google Scholar] [CrossRef] [PubMed]
  9. Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803. [Google Scholar] [CrossRef] [PubMed]
  10. Musarra-Pizzo, M.; Pennisi, R.; Ben-Amor, I.; Mandalari, G.; Sciortino, M.T. Antiviral Activity Exerted by Natural Products against Human Viruses. Viruses 2021, 13, 828. [Google Scholar] [CrossRef]
  11. Lin, L.-T.; Hsu, W.-C.; Lin, C.-C. Antiviral Natural Products and Herbal Medicines. J. Tradit. Complement. Med. 2014, 4, 24–35. [Google Scholar] [CrossRef] [PubMed]
  12. Stan, D.; Enciu, A.-M.; Mateescu, A.L.; Ion, A.C.; Brezeanu, A.C.; Stan, D.; Tanase, C. Natural Compounds With Antimicrobial and Antiviral Effect and Nanocarriers Used for Their Transportation. Front. Pharmacol. 2021, 12, 723233. [Google Scholar] [CrossRef] [PubMed]
  13. Shamshirgaran, M.; Maleki, A.; Askari, P.; Yousefi, M.; Malaki Moghadam, H.; Aramjoo, H.; Zare_Bidaki, M. Antibacterial Effects of the Aqueous Extract of Lycopersicon Esculentum Mill Native in South Khorasan of Iran against Four Species Associated with Gastrointestinal Infections. J. Basic Res. Med. Sci. 2020, 7, 1–6. [Google Scholar]
  14. Heber, D.; Lu, Q.-Y. Overview of Mechanisms of Action of Lycopene. Exp. Biol. Med. 2002, 227, 920–923. [Google Scholar] [CrossRef] [PubMed]
  15. Tilahun, S.; Park, D.S.; Seo, M.H.; Jeong, C.S. Review on Factors Affecting the Quality and Antioxidant Properties of Tomatoes. AJB 2017, 16, 1678–1687. [Google Scholar] [CrossRef]
  16. Kumar, M.; Tomar, M.; Bhuyan, D.J.; Punia, S.; Grasso, S.; Sá, A.G.A.; Carciofi, B.A.M.; Arrutia, F.; Changan, S.; Singh, S.; et al. Tomato (Solanum lycopersicum L.) Seed: A Review on Bioactives and Biomedical Activities. Biomed. Pharmacother. 2021, 142, 112018. [Google Scholar] [CrossRef]
  17. Nakayasu, M.; Takamatsu, K.; Yazaki, K.; Sugiyama, A. Plant Specialized Metabolites in the Rhizosphere of Tomatoes: Secretion and Effects on Microorganisms. Biosci. Biotechnol. Biochem. 2022, 87, 13–20. [Google Scholar] [CrossRef]
  18. Kamilova, F.; Kravchenko, L.V.; Shaposhnikov, A.I.; Azarova, T.; Makarova, N.; Lugtenberg, B. Organic Acids, Sugars, and l-Tryptophane in Exudates of Vegetables Growing on Stonewool and Their Effects on Activities of Rhizosphere Bacteria. MPMI 2006, 19, 250–256. [Google Scholar] [CrossRef]
  19. Simons, M.; Permentier, H.P.; de Weger, L.A.; Wijffelman, C.A.; Lugtenberg, B.J.J. Amino Acid Synthesis Is Necessary for Tomato Root Colonization by Pseudomonas Fluorescens Strain WCS365. MPMI 1997, 10, 102–106. [Google Scholar] [CrossRef]
  20. Zboralski, A.; Filion, M. Genetic Factors Involved in Rhizosphere Colonization by Phytobeneficial Pseudomonas Spp. Comput. Struct. Biotechnol. J. 2020, 18, 3539–3554. [Google Scholar] [CrossRef]
  21. Sahlin, E.; Savage, G.P.; Lister, C.E. Investigation of the Antioxidant Properties of Tomatoes after Processing. J. Food Compos. Anal. 2004, 17, 635–647. [Google Scholar] [CrossRef]
  22. Miller, E.C.; Giovannucci, E.; Erdman, J.W.; Bahnson, R.; Schwartz, S.J.; Clinton, S.K. Tomato Products, Lycopene, and Prostate Cancer Risk. Urol. Clin. N. Am. 2002, 29, 83–93. [Google Scholar] [CrossRef] [PubMed]
  23. Story, E.N.; Kopec, R.E.; Schwartz, S.J.; Harris, G.K. An Update on the Health Effects of Tomato Lycopene. Annu. Rev. Food Sci. Technol. 2010, 1, 189–210. [Google Scholar] [CrossRef] [PubMed]
  24. Diosa-Toro, M.; Troost, B.; van de Pol, D.; Heberle, A.M.; Urcuqui-Inchima, S.; Thedieck, K.; Smit, J.M. Tomatidine, a Novel Antiviral Compound towards Dengue Virus. Antivir. Res. 2019, 161, 90–99. [Google Scholar] [CrossRef] [PubMed]
  25. Troost, B.; Mulder, L.M.; Diosa-Toro, M.; van de Pol, D.; Rodenhuis-Zybert, I.A.; Smit, J.M. Tomatidine, a Natural Steroidal Alkaloid Shows Antiviral Activity towards Chikungunya Virus in Vitro. Sci. Rep. 2020, 10, 6364. [Google Scholar] [CrossRef] [PubMed]
  26. Thorne, H.V.; Clarke, G.F.; Skuce, R. The Inactivation of Herpes Simplex Virus by Some Solanaceae Glycoalkaloids. Antivir. Res. 1985, 5, 335–343. [Google Scholar] [CrossRef] [PubMed]
  27. Santoro, V.; Schiavon, M.; Visentin, I.; Constán-Aguilar, C.; Cardinale, F.; Celi, L. Strigolactones Affect Phosphorus Acquisition Strategies in Tomato Plants. Plant Cell Environ. 2021, 44, 3628–3642. [Google Scholar] [CrossRef]
  28. Pasquero, S.; Gugliesi, F.; Biolatti, M.; Dell’Oste, V.; Albano, C.; Bajetto, G.; Griffante, G.; Trifirò, L.; Brugo, B.; Raviola, S.; et al. Citrullination Profile Analysis Reveals Peptidylarginine Deaminase 3 as an HSV-1 Target to Dampen the Activity of Candidate Antiviral Restriction Factors. PLoS Pathog. 2023, 19, e1011849. [Google Scholar] [CrossRef]
  29. Biolatti, M.; Blangetti, M.; D’Arrigo, G.; Spyrakis, F.; Cappello, P.; Albano, C.; Ravanini, P.; Landolfo, S.; De Andrea, M.; Prandi, C.; et al. Strigolactone Analogs Are Promising Antiviral Agents for the Treatment of Human Cytomegalovirus Infection. Microorganisms 2020, 8, E703. [Google Scholar] [CrossRef]
  30. Biolatti, M.; Blangetti, M.; Baggieri, M.; Marchi, A.; Gioacchini, S.; Bajetto, G.; Arnodo, D.; Bucci, P.; Fioravanti, R.; Kojouri, M.; et al. Strigolactones as Broad-Spectrum Antivirals against β-Coronaviruses through Targeting the Main Protease Mpro. ACS Infect. Dis. 2023, 7, 1310–1318. [Google Scholar] [CrossRef]
  31. Sprague, E.R.; Reinhard, H.; Cheung, E.J.; Farley, A.H.; Trujillo, R.D.; Hengel, H.; Bjorkman, P.J. The Human Cytomegalovirus Fc Receptor gp68 Binds the Fc CH2-CH3 Interface of Immunoglobulin G. J. Virol. 2008, 82, 3490–3499. [Google Scholar] [CrossRef] [PubMed]
  32. Cole, S. Herpes Simplex Virus: Epidemiology, Diagnosis, and Treatment. Nurs. Clin. N. Am. 2020, 55, 337–345. [Google Scholar] [CrossRef] [PubMed]
  33. Whitley, R.J.; Roizman, B. Herpes Simplex Virus Infections. Lancet 2001, 357, 1513–1518. [Google Scholar] [CrossRef] [PubMed]
  34. Kumar, A.; De, S.; Moharana, A.K.; Nayak, T.K.; Saswat, T.; Datey, A.; Mamidi, P.; Mishra, P.; Subudhi, B.B.; Chattopadhyay, S. Inhibition of Herpes Simplex Virus-1 Infection by MBZM-N-IBT: In Silico and in Vitro Studies. Virol. J. 2021, 18, 103. [Google Scholar] [CrossRef] [PubMed]
  35. Gentry, B.G.; Drach, J.C. Metabolism of Cyclopropavir and Ganciclovir in Human Cytomegalovirus-Infected Cells. Antimicrob. Agents Chemother. 2014, 58, 2329–2333. [Google Scholar] [CrossRef]
  36. Ikeda, T.; Ando, J.; Miyazono, A.; Zhu, X.H.; Tsumagari, H.; Nohara, T.; Yokomizo, K.; Uyeda, M. Anti-Herpes Virus Activity of Solanum Steroidal Glycosides. Biol. Pharm. Bull. 2000, 23, 363–364. [Google Scholar] [CrossRef]
  37. Valadares, Y.M.; Brandão’a, G.C.; Kroon, E.G.; Filho, J.D.S.; Oliveira, A.B.; Braga, F.C. Antiviral Activity of Solanum Paniculatum Extract and Constituents. Z. Naturforsch C J. Biosci. 2009, 64, 813–818. [Google Scholar] [CrossRef]
  38. Kumar, B.R. Application of HPLC and ESI-MS Techniques in the Analysis of Phenolic Acids and Flavonoids from Green Leafy Vegetables (GLVs). J. Pharm. Anal. 2017, 7, 349–364. [Google Scholar] [CrossRef]
  39. Ali, M.Y.; Sina, A.A.I.; Khandker, S.S.; Neesa, L.; Tanvir, E.M.; Kabir, A.; Khalil, M.I.; Gan, S.H. Nutritional Composition and Bioactive Compounds in Tomatoes and Their Impact on Human Health and Disease: A Review. Foods 2020, 10, 45. [Google Scholar] [CrossRef] [PubMed]
  40. Moco, S.; Bino, R.J.; Vorst, O.; Verhoeven, H.A.; de Groot, J.; van Beek, T.A.; Vervoort, J.; de Vos, C.H.R. A Liquid Chromatography-Mass Spectrometry-Based Metabolome Database for Tomato. Plant Physiol. 2006, 141, 1205–1218. [Google Scholar] [CrossRef] [PubMed]
  41. Sánchez-Fidalgo, S.; Villegas, I.; Aparicio-Soto, M.; Cárdeno, A.; Rosillo, M.Á.; González-Benjumea, A.; Marset, A.; López, Ó.; Maya, I.; Fernández-Bolaños, J.G.; et al. Effects of Dietary Virgin Olive Oil Polyphenols: Hydroxytyrosyl Acetate and 3, 4-Dihydroxyphenylglycol on DSS-Induced Acute Colitis in Mice. J. Nutr. Biochem. 2015, 26, 513–520. [Google Scholar] [CrossRef]
  42. Toujani, M.M.; Rittà, M.; Civra, A.; Genovese, S.; Epifano, F.; Ghram, A.; Lembo, D.; Donalisio, M. Inhibition of HSV-2 Infection by Pure Compounds from Thymus Capitatus Extract in Vitro. Phytother. Res. 2018, 32, 1555–1563. [Google Scholar] [CrossRef]
  43. Zakaryan, H.; Arabyan, E.; Oo, A.; Zandi, K. Flavonoids: Promising Natural Compounds against Viral Infections. Arch. Virol. 2017, 162, 2539–2551. [Google Scholar] [CrossRef]
  44. Lyu, S.-Y.; Rhim, J.-Y.; Park, W.-B. Antiherpetic Activities of Flavonoids against Herpes Simplex Virus Type 1 (HSV-1) and Type 2 (HSV-2) in Vitro. Arch. Pharm. Res. 2005, 28, 1293–1301. [Google Scholar] [CrossRef] [PubMed]
  45. Lee, S.; Lee, H.H.; Shin, Y.S.; Kang, H.; Cho, H. The Anti-HSV-1 Effect of Quercetin Is Dependent on the Suppression of TLR-3 in Raw 264.7 Cells. Arch. Pharm. Res. 2017, 40, 623–630. [Google Scholar] [CrossRef]
  46. Hung, P.-Y.; Ho, B.-C.; Lee, S.-Y.; Chang, S.-Y.; Kao, C.-L.; Lee, S.-S.; Lee, C.-N. Houttuynia Cordata Targets the Beginning Stage of Herpes Simplex Virus Infection. PLoS ONE 2015, 10, e0115475. [Google Scholar] [CrossRef] [PubMed]
  47. Periferakis, A.; Periferakis, A.-T.; Troumpata, L.; Periferakis, K.; Scheau, A.-E.; Savulescu-Fiedler, I.; Caruntu, A.; Badarau, I.A.; Caruntu, C.; Scheau, C. Kaempferol: A Review of Current Evidence of Its Antiviral Potential. Int. J. Mol. Sci. 2023, 24, 16299. [Google Scholar] [CrossRef] [PubMed]
  48. Hossen, M.S.; Ali, M.Y.; Jahurul, M.H.A.; Abdel-Daim, M.M.; Gan, S.H.; Khalil, M.I. Beneficial Roles of Honey Polyphenols against Some Human Degenerative Diseases: A Review. Pharmacol. Rep. 2017, 69, 1194–1205. [Google Scholar] [CrossRef] [PubMed]
  49. Agarwal, S.; Rao, A.V. Tomato Lycopene and Its Role in Human Health and Chronic Diseases. CMAJ 2000, 163, 739–744. [Google Scholar] [PubMed]
  50. Navarro-González, I.; García-Alonso, J.; Periago, M.J. Bioactive Compounds of Tomato: Cancer Chemopreventive Effects and Influence on the Transcriptome in Hepatocytes. J. Funct. Foods 2018, 42, 271–280. [Google Scholar] [CrossRef]
  51. Imran, M.; Ghorat, F.; Ul-Haq, I.; Ur-Rehman, H.; Aslam, F.; Heydari, M.; Shariati, M.A.; Okuskhanova, E.; Yessimbekov, Z.; Thiruvengadam, M.; et al. Lycopene as a Natural Antioxidant Used to Prevent Human Health Disorders. Antioxidants 2020, 9, 706. [Google Scholar] [CrossRef]
  52. Zhu, R.; Chen, B.; Bai, Y.; Miao, T.; Rui, L.; Zhang, H.; Xia, B.; Li, Y.; Gao, S.; Wang, X.-D.; et al. Lycopene in Protection against Obesity and Diabetes: A Mechanistic Review. Pharmacol. Res. 2020, 159, 104966. [Google Scholar] [CrossRef]
  53. Saini, R.K.; Rengasamy, K.R.R.; Mahomoodally, F.M.; Keum, Y.-S. Protective Effects of Lycopene in Cancer, Cardiovascular, and Neurodegenerative Diseases: An Update on Epidemiological and Mechanistic Perspectives. Pharmacol. Res. 2020, 155, 104730. [Google Scholar] [CrossRef] [PubMed]
  54. Sakemi, Y.; Sato, K.; Hara, K.; Honda, M.; Shindo, K. Biological Activities of Z-Lycopenes Contained in Food. J. Oleo Sci. 2020, 69, 1509–1516. [Google Scholar] [CrossRef] [PubMed]
  55. Zhang, Z.; Li, Q.; Liu, F.; Wang, D. Lycoperoside H Protects against Diabetic Nephropathy via Alteration of Gut Microbiota and Inflammation. J. Biochem. Mol. Toxicol. 2022, 36, e23216. [Google Scholar] [CrossRef] [PubMed]
  56. Thomas, E.; Stewart, L.E.; Darley, B.A.; Pham, A.M.; Esteban, I.; Panda, S.S. Plant-Based Natural Products and Extracts: Potential Source to Develop New Antiviral Drug Candidates. Molecules 2021, 26, 6197. [Google Scholar] [CrossRef]
Figure 1. Strategy for the preparation of extracts from tomato root exudates (created with BioRender).
Figure 1. Strategy for the preparation of extracts from tomato root exudates (created with BioRender).
Microorganisms 12 00373 g001
Figure 2. Antiviral activity of tomato extract against HSV-1. (A) VERO cells were treated with different concentrations of the exudate or the same amount of vehicle solution (CTRL). Forty-eight hours after treatment, cell viability was determined using the MTT assay. Data are expressed as mean value percent cell viability vs. vehicle control cells (set at 100%). Bars represent the means ± SEM from three independent experiments. (B) VERO cells were pre-treated with serial concentrations of tomato extract or CTRL for 1 h. Subsequently, cells were infected with HSV-1 (MOI 0.1), and after the virus adsorption, the viral inoculum was removed; cultures were exposed to the extract or ACV during the infection and for 48 h thereafter. The extent of HSV-1 replication was then assessed by titrating the infectivity of the cell extracts and supernatants combined using a standard plaque assay. Histograms were obtained by plotting the mean plaque counts for each dilution expressed as PFU/mL. Bars represent the means and ± SEM from five independent experiments (p < 0.05 *, p < 0.0001 ****, unpaired t-test tomato extract vs. CTRL). (C) VERO cells were treated with the exudate (400 µg/mL) or with an equal volume of CTRL 1 h before infection and for the entire duration of the infection and infected with HSV-1 at an MOI of 1. Samples were collected at the indicated time points, lysed, and subjected to Western blot analyses. ACV was used as a reference drug. Membranes were stained with the indicated antibodies. (D) VERO cells were treated with the tomato extract (400 µg/mL) or with the same volume of CTRL and infected with HSV-1 (MOI 1). After 24 h, DNA was extracted from infected cells, and the number of HSV-1 genomes was quantified by qPCR. To determine the number of viral DNA genomes, the primers were used to amplify a segment of the gE gene, and the cellular housekeeping gene GAPDH was used to normalize viral genome counts. Bars represent the means ± SEM from three independent experiments (p < 0.01 **, unpaired t-test tomato extract vs. CTRL).
Figure 2. Antiviral activity of tomato extract against HSV-1. (A) VERO cells were treated with different concentrations of the exudate or the same amount of vehicle solution (CTRL). Forty-eight hours after treatment, cell viability was determined using the MTT assay. Data are expressed as mean value percent cell viability vs. vehicle control cells (set at 100%). Bars represent the means ± SEM from three independent experiments. (B) VERO cells were pre-treated with serial concentrations of tomato extract or CTRL for 1 h. Subsequently, cells were infected with HSV-1 (MOI 0.1), and after the virus adsorption, the viral inoculum was removed; cultures were exposed to the extract or ACV during the infection and for 48 h thereafter. The extent of HSV-1 replication was then assessed by titrating the infectivity of the cell extracts and supernatants combined using a standard plaque assay. Histograms were obtained by plotting the mean plaque counts for each dilution expressed as PFU/mL. Bars represent the means and ± SEM from five independent experiments (p < 0.05 *, p < 0.0001 ****, unpaired t-test tomato extract vs. CTRL). (C) VERO cells were treated with the exudate (400 µg/mL) or with an equal volume of CTRL 1 h before infection and for the entire duration of the infection and infected with HSV-1 at an MOI of 1. Samples were collected at the indicated time points, lysed, and subjected to Western blot analyses. ACV was used as a reference drug. Membranes were stained with the indicated antibodies. (D) VERO cells were treated with the tomato extract (400 µg/mL) or with the same volume of CTRL and infected with HSV-1 (MOI 1). After 24 h, DNA was extracted from infected cells, and the number of HSV-1 genomes was quantified by qPCR. To determine the number of viral DNA genomes, the primers were used to amplify a segment of the gE gene, and the cellular housekeeping gene GAPDH was used to normalize viral genome counts. Bars represent the means ± SEM from three independent experiments (p < 0.01 **, unpaired t-test tomato extract vs. CTRL).
Microorganisms 12 00373 g002
Figure 3. Antiviral activity of the tomato extract against HCMV. (A) HFF cells were treated with different concentrations of the exudate or with the same amount of vehicle solution (CTRL). After six days, cell viability was determined using the MTT assay. Data are expressed as mean value percent cell viability vs. CTRL (set at 100%). Bars represent the means ± SEM from three independent experiments. (B) HFF cells were pre-treated with serial concentrations of tomato extract for 1 h. Subsequently, cells were infected with HCMV (MOI 0.1), and after the virus adsorption, the viral inoculum was removed; cultures were exposed to the extract, GCV, or CTRL during the infection and for 144 h thereafter. The extent of HCMV replication was then assessed by titrating the infectivity of cell extract and supernatants combined using a standard plaque assay. Histograms were obtained by plotting the mean plaque counts for each dilution expressed as PFU/mL. Bars represent the means ± SEM from five independent experiments (p < 0.01 **, p < 0.001 ***, unpaired t-test tomato extract vs. CTRL). (C) HFF cells were treated with tomato extract (100 µg/mL) or with an equal volume of vehicle 1 h before infection and for the entire duration of the infection and infected with HCMV at an MOI of 1. Samples were collected at the indicated time points, lysed, and subjected to Western blot analyses. GCV was used as a reference drug. Membranes were stained with the indicated antibodies. (D) HFF cells were treated with the root extract (100 µg/mL) or with the same volume of CTRL and infected with HCMV (MOI 1). After 72 h, DNA was extracted, and the number of HCMV genomes was quantified by qPCR. To determine the number of viral DNA genomes, the primers were used to amplify a segment of the IE1 gene, and the cellular housekeeping gene GAPDH was used to normalize viral genome counts. Bars represent the means ± SEM from three independent experiments (p < 0.05 *, unpaired t-test tomato extract vs. CTRL).
Figure 3. Antiviral activity of the tomato extract against HCMV. (A) HFF cells were treated with different concentrations of the exudate or with the same amount of vehicle solution (CTRL). After six days, cell viability was determined using the MTT assay. Data are expressed as mean value percent cell viability vs. CTRL (set at 100%). Bars represent the means ± SEM from three independent experiments. (B) HFF cells were pre-treated with serial concentrations of tomato extract for 1 h. Subsequently, cells were infected with HCMV (MOI 0.1), and after the virus adsorption, the viral inoculum was removed; cultures were exposed to the extract, GCV, or CTRL during the infection and for 144 h thereafter. The extent of HCMV replication was then assessed by titrating the infectivity of cell extract and supernatants combined using a standard plaque assay. Histograms were obtained by plotting the mean plaque counts for each dilution expressed as PFU/mL. Bars represent the means ± SEM from five independent experiments (p < 0.01 **, p < 0.001 ***, unpaired t-test tomato extract vs. CTRL). (C) HFF cells were treated with tomato extract (100 µg/mL) or with an equal volume of vehicle 1 h before infection and for the entire duration of the infection and infected with HCMV at an MOI of 1. Samples were collected at the indicated time points, lysed, and subjected to Western blot analyses. GCV was used as a reference drug. Membranes were stained with the indicated antibodies. (D) HFF cells were treated with the root extract (100 µg/mL) or with the same volume of CTRL and infected with HCMV (MOI 1). After 72 h, DNA was extracted, and the number of HCMV genomes was quantified by qPCR. To determine the number of viral DNA genomes, the primers were used to amplify a segment of the IE1 gene, and the cellular housekeeping gene GAPDH was used to normalize viral genome counts. Bars represent the means ± SEM from three independent experiments (p < 0.05 *, unpaired t-test tomato extract vs. CTRL).
Microorganisms 12 00373 g003
Figure 4. Time of addition assay. VERO or HFF cells were (A) pretreated with serial dilutions of tomato extract for 1 h, after which, treatments were removed and cells were infected with HSV-1 (VERO) or HCMV (HFF) (MOI 0.1); (B) infected and treated simultaneously for 2 h, after which treatments and viral inoculum were removed; (C) infected as described above and, after the removal of inoculum, treated with increasing concentrations of the compound. For all the conditions, cells were incubated for 48 h (VERO) or 144 h (HFF); then, samples were harvested, and infectivity titers were determined by a standard plaque assay. Histograms were obtained by plotting the mean plaque counts for each dilution expressed as PFU/mL. Bars represent the means ± SEM from three independent experiments (p < 0.05 *, p < 0.01 **, p < 0.001 ***, p < 0.0001 ****, unpaired t-test tomato extract vs. CTRL).
Figure 4. Time of addition assay. VERO or HFF cells were (A) pretreated with serial dilutions of tomato extract for 1 h, after which, treatments were removed and cells were infected with HSV-1 (VERO) or HCMV (HFF) (MOI 0.1); (B) infected and treated simultaneously for 2 h, after which treatments and viral inoculum were removed; (C) infected as described above and, after the removal of inoculum, treated with increasing concentrations of the compound. For all the conditions, cells were incubated for 48 h (VERO) or 144 h (HFF); then, samples were harvested, and infectivity titers were determined by a standard plaque assay. Histograms were obtained by plotting the mean plaque counts for each dilution expressed as PFU/mL. Bars represent the means ± SEM from three independent experiments (p < 0.05 *, p < 0.01 **, p < 0.001 ***, p < 0.0001 ****, unpaired t-test tomato extract vs. CTRL).
Microorganisms 12 00373 g004
Figure 5. Full-scan positive ion ESI-MS spectrum of a solution of tomato root exudate. Solution infused: 1.0 mg/mL in acetonitrile + 0.1% formic acid.
Figure 5. Full-scan positive ion ESI-MS spectrum of a solution of tomato root exudate. Solution infused: 1.0 mg/mL in acetonitrile + 0.1% formic acid.
Microorganisms 12 00373 g005
Table 1. Identification of phytochemicals of tomato root exudate components. Analytically significant ions (m/z) are reported.
Table 1. Identification of phytochemicals of tomato root exudate components. Analytically significant ions (m/z) are reported.
CompoundMolecular
Formula
M.W.
(g/mol)
Ionized
Adduct
Signal
(m/z)
LycopeneC40H56536.88[M + H]+537
β-CaroteneC40H56536.88[M + H]+537
α-TomatineC50H83NO211034.18[M + H + Na]2+529
Lycoperoside HC50H83NO221049.54[M + 2H]2+ 525
β-SitosterolC29H50O414.72[M + H]+416
TomatidineC27H45NO2415.66[M + H]+416
GlcNAc-(1→N)-AsnC12H21N3O8335.31[M + NH4]+353
MyricetinC15H10O8318.04[M + NH4]+
[M + H + NH4]2+
336 a
168
IsorhamnetinC16H12O7316.26[M + NH4]+334
QuercetinC15H10O7302.24[M + NH4]+320
LuteinC40H56O2568.87[M + H + Na]2+296
KaempferolC15H10O6286.23[M + H]+287 a
PhytoeneC40H64544.95[M + H + NH4]2+282
PhytoflueneC40H62542.94[M + H + NH4]2+280 a
AHYPC7H11NO4173.17[M + H]+174 a
HomocysteineC4H9NO2S135.18[M + H]+136
a Low-abundance ions.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bajetto, G.; Arnodo, D.; Biolatti, M.; Trifirò, L.; Albano, C.; Pasquero, S.; Gugliesi, F.; Campo, E.; Spyrakis, F.; Prandi, C.; et al. Antiherpetic Activity of a Root Exudate from Solanum lycopersicum. Microorganisms 2024, 12, 373. https://doi.org/10.3390/microorganisms12020373

AMA Style

Bajetto G, Arnodo D, Biolatti M, Trifirò L, Albano C, Pasquero S, Gugliesi F, Campo E, Spyrakis F, Prandi C, et al. Antiherpetic Activity of a Root Exudate from Solanum lycopersicum. Microorganisms. 2024; 12(2):373. https://doi.org/10.3390/microorganisms12020373

Chicago/Turabian Style

Bajetto, Greta, Davide Arnodo, Matteo Biolatti, Linda Trifirò, Camilla Albano, Selina Pasquero, Francesca Gugliesi, Eva Campo, Francesca Spyrakis, Cristina Prandi, and et al. 2024. "Antiherpetic Activity of a Root Exudate from Solanum lycopersicum" Microorganisms 12, no. 2: 373. https://doi.org/10.3390/microorganisms12020373

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop