Next Article in Journal
Monitoring Viral Entry in Real-Time Using a Luciferase Recombinant Vesicular Stomatitis Virus Producing SARS-CoV-2, EBOV, LASV, CHIKV, and VSV Glycoproteins
Next Article in Special Issue
Chloroquine and Sulfadoxine Derivatives Inhibit ZIKV Replication in Cervical Cells
Previous Article in Journal
Application of a Sanger-Based External Quality Assurance Strategy for the Transition of HIV-1 Drug Resistance Assays to Next Generation Sequencing
Previous Article in Special Issue
IMU-838, a Developmental DHODH Inhibitor in Phase II for Autoimmune Disease, Shows Anti-SARS-CoV-2 and Broad-Spectrum Antiviral Efficacy In Vitro
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Resveratrol, Rapamycin and Metformin as Modulators of Antiviral Pathways

by
Francesca Benedetti
1,†,
Vincenzo Sorrenti
2,3,4,†,
Alessandro Buriani
4,
Stefano Fortinguerra
5,
Giovanni Scapagnini
6,* and
Davide Zella
1,*
1
Institute of Human Virology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
2
Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
3
Bendessere™ Study Center, Via Prima Strada 23/3, 35129 Padova, Italy
4
Maria Paola Belloni Center for Personalized Medicine, Data Medica Group (Synlab Limited), 35100 Padova, Italy
5
IRCCS SDN, 80143 Napoli, Italy
6
Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
*
Authors to whom correspondence should be addressed.
Authors contributed equally.
Submission received: 11 November 2020 / Revised: 14 December 2020 / Accepted: 15 December 2020 / Published: 17 December 2020
(This article belongs to the Special Issue Unconventional Antiviral Agents)

Abstract

:
Balanced nutrition and appropriate dietary interventions are fundamental in the prevention and management of viral infections. Additionally, accurate modulation of the inflammatory response is necessary to achieve an adequate antiviral immune response. Many studies, both in vitro with mammalian cells and in vivo with small animal models, have highlighted the antiviral properties of resveratrol, rapamycin and metformin. The current review outlines the mechanisms of action of these three important compounds on the cellular pathways involved with viral replication and the mechanisms of virus-related diseases, as well as the current status of their clinical use.

1. Resveratrol

Resveratrol (trans-3,4,5-trihydroxystilbene) is a small polyphenol natural molecule that can be obtained from several sources, including red wine, grapes, a variety of berries, peanuts and certain medicinal plants [1,2]. With lipophilic characteristics that make it easily absorbed, metabolized and excreted, resveratrol is used as a dietary supplement, even though it has a low bioavailability that limits its use [3,4,5]. Indeed, the way it is consumed, and the kind of food ingested influence its absorption [6]. Upon oral administration, resveratrol complexes with membrane transporters, which allow its absorption by passive diffusion, eventually ending up in the bloodstream, where it can be detected in three forms: glucuronide, sulfate and free [6]. The latter form is typically associated with albumin or lipoproteins, to allow for better circulation and entry in the cells [7]. Further metabolism of resveratrol takes place in the liver, producing different metabolites that can be detected in the urine [8].
Both in vitro experiments with mammalian cells and in vivo experiments with small animal models have shown a number of positive effects correlated with the use of resveratrol [9,10,11,12], including reduction of inflammation [13], antioxidant and antiaging [14,15,16], and anticancer [17,18,19,20,21]. For this reason, resveratrol is considered a promising molecule for the prevention and treatment of a number of conditions, including autoimmune disorders, cardiovascular and neurodegenerative diseases as well as some chronic diseases [22,23,24,25,26]. In addition, resveratrol was demonstrated to extend the lifespan of several evolutionarily distant species, including Saccharomyces cerevisiae [27], Caenorhabditis elegans [28] and Drosophila melanogaster [29], thus further confirming the conserved nature of the pathways involved.
A number of in vivo studies have provided the rationale for the use of the polyphenolic molecule resveratrol in several humans disorders. In particular, inhibition of important gene pathways like the NF-κB pathway supports its use as an antioxidant, while inhibitions of viral replication, protein synthesis, gene expression and nucleic acid synthesis support its use as an antiviral [30]. In fact, resveratrol is considered a molecule with promising immunomodulatory effects which is able to regulate immunological and inflammatory responses.
A number of human viruses seem to be sensitive to the effects of resveratrol (Table 1), including influenza virus [31], respiratory syncytial virus [32,33,34,35,36,37], varicella zoster virus [38], Epstein-Barr virus [39,40], herpes simplex virus [41,42,43,44], human immunodeficiency virus [45,46,47] and enterovirus [48,49]. While in these cases, treatment with resveratrol reduced viral infection, a notable exception was an in vitro model of Hepatitis C where infection was enhanced [50].
Using the mammalian MDCK cells normally used for influenza research as in vitro model, Palamara et al. (2005) showed that resveratrol treatment resulted in inhibition of viral nucleoproteins translocation to the nucleus, and that this diminished the inhibitory effect of protein kinase C related pathways. The effect was not correlated with the glutathione-mediated antioxidant activity of resveratrol [31].
Regarding respiratory syncytial virus (RPSV) infection, a disease of the respiratory system, in vivo treatment of mice (injected intraperitoneally with resveratrol 30 mg/Kg/day for 5 days) resulted in amelioration of inflammatory symptoms, likely because of modulation of several molecules, including induction of muscarinic 2 receptor (M2R), inhibition of toll/IL-1R domain-containing adaptor inducing IFN (TRIF) signaling and reduction of toll-like receptor 3 expression [36]. In yet another set of experiments in mice (injected intraperitoneally with resveratrol 30 mg/Kg/day for five days), experimental data showed decreased IFN-γ expression accompanied by reduced airway inflammation and decreased hyper-responsiveness (AHR). A number of other proteins were also affected, including: (i) increased expression of sterile-α- and armadillo motif-containing protein (SARM), (ii) decreased expression of matrix metalloproteinase 12 (MMP-12) and, (iii) decreased TIR-domain-containing adapter-inducing interferon-β (TRIF) [33,51]. Additionally, in vitro studies, where resveratrol was used to treat epithelial cells infected with RPSV, showed reduction in both production of interleukin- (IL-) 6 and in viral replication. Moreover, hampering of the expression of viral induced toll-like receptor domain and TANK binding kinase 1 (TBK1) protein [34] was observed. Finally, resveratrol in combination with a baicalin (a flavonoid found in numerous species of Scutellaria) joint enema (resveratrol administered 30 mg/Kg/day retention enema for 30 min) increased the antiviral mechanisms in mice infected with RPSV, namely increasing the levels of IL-2, IFN-γ, and tumor necrosis factor-alpha (TNF-α) [32].
With respect to Varicella Zooster Virus (VZV), resveratrol noticeably decreased the synthesis of intermediate early protein (IE 62) in MRC-5 cells (human lung cells line) early in infection (within 30 h) [38].
Resveratrol inhibited early antigen production of Epstein-Barr virus (EBV) in infected B-cells (Raji) [53]. Together with decreasing EBV lytic cycle by reducing transcription genes and proteins, (Rta, Zta, and diffused early antigen, EA-D), as well as hampering the activity by EBV immediate-early proteins (BRLF1 and BZLF1 promoters), resveratrol reduced the production of viral particles [40]. These data confirmed early findings showing dose-dependent inhibition by resveratrol of expression of lytic genes and viral particles, where its major antiviral mechanism was associated with (i) inhibition of transcription factors NF-κB and AP1; (ii) inhibition of protein synthesis, and (iii) reduction in ROS production [53]. Due to the broad recognition of EBV as an oncogenic virus, resveratrol was employed to prevent transformation of human B-cells caused by EBV. The effect of resveratrol was mainly observed on the downregulation of two antiapoptotic proteins, namely Mc1 and survivin, together with the suppression of signaling induced by NF-κB, STAT-3, miR-155 and miR-34a in EBV infected cells [39]. Finally, resveratrol (at a concentration of 85 nM over the duration of the experiment, administered by topic treatment at the site of skin area where tumorigenesis was promoted) was shown to reduce papilloma production by 60% in EBV-infected mice 20 weeks post inoculation [52].
Treatment with resveratrol resulted in the decreased production of early viral protein ICP-4 of herpes simplex virus-1 and herpes simplex virus-2 (HSV-1 and HSV-2) in Vero cells (kidney cells derived from monkey) and infected human lung cells (MRC-5). Consequently, the viral replication and load was reduced in a dose-dependent and reversible manner [42]. In addition, treatment with resveratrol of latently infected neuron cells deferred the interphase stage of the cell cycle and hindered reactivation of HSVs [43]. In in vivo studies on nude mice, a topical unguent of 12.5% and 15% of resveratrol applied 2–5 times daily for five days on scratched skin infected with HSV-1 inhibited the development of cutaneous lesions [44]. Moreover, a vaginal cream of 19% resveratrol applied topically to mice after local infection with HSV-2 and HSV-1 prevented the development of vaginal lesions with a 3% mortality rate, as opposed to the 37% mortality rate observed in the placebo group [42]. Induction of a quick and continuous release of ROS, resulting in the inhibition of NF-κB and extracellular signal-regulated kinases/mitogen-activated protein kinases (ERK/MAPK), together with a marked reduction in the expression of several HSV genes (immediate-early, early, and late) and viral DNA synthesis were the most likely molecular mechanisms ascribed to these effects induced by resveratrol [41,54].
Resveratrol and its analogs, alone or in combination with decitabine (a nucleoside used for myelodysplastic syndromes), has been employed in vitro (5 μM) to inhibit DNA synthesis of the HIV molecular clone NL4–3 (which harbors the mutant M184V reverse transcriptase (RT)) during the reverse transcription step of the HIV life cycle [45,46]. To further support the possibility of using resveratrol (or its analogues) as adjuvant in anti-HIV preexposure prophylaxis (PrEP) formulation, it was shown that low doses of resveratrol (and its chemical relative pterostilbene) were able to block HIV-1 infection in resting CD4 T cells, interfering with the step involving reverse transcription [47]. This anti-HIV effect was abrogated in the presence of Vpx, an HIV-1 and simian immunodeficiency virus protein that causes an increase of deoxynucleoside triphosphate (dNTP) levels, or when exogenous deoxynucleosides where added to the culture medium. These findings further confirm the ability of resveratrol to inhibit ribonucleotide reductase and to decrease dNTP levels in cells. Another resveratrol analog, 3,3′,4,4′,5,5′-hexahydroxy-trans-stilbene (M8), was shown to have a potent anti-HIV activity against different viral variants. More in detail, M8 blocked HIV fusion with host cells, indicating an effect at the very early steps of viral entry. Taken together, these latest data indicate that the novel resveratrol derivative, M8, has anti-HIV-1 effects and likely a different mechanism of action than anti-HIV-1 drugs currently in use [55].
Resveratrol treatment also inhibited the synthesis of Enterovirus 71 (EV 71) viral protein 1 (VP1) and phosphorylation of proinflammatory cytokines (IKKα, IKKβ, IKKγ, IKBα, NF-κB p50, and NF-κB p65) in Rhabdomyosarcoma cell line, together with secretion of IL-6 and TNF-α [48].
In Vero E6 cells infected by MERS-CoV, resveratrol treatment downregulated the apoptosis induced by viral infection which resulted in increased survival of infected cells, and reduced the expression of nucleocapsid (N) protein, essential for MERS-CoV replication [56]. The use of resveratrol has also been proposed in the treatment of patients infected with SARS-CoV-2, due to its ability to decrease the high levels of inflammatory cytokines, such as IL-6 and TNF-α, observed in patients [58,59]. Further data indicating a potential beneficial effect of resveratrol in the treatment of SARS-CoV-2 patients come from the observation that upregulation of Angiotensin Converting Enzyme-2 (ACE-2) has a protective effect on SARS-CoV illness severity, and high intake of resveratrol upregulates ACE-2 receptors [57,60].
A number of molecules have been identified as targets for resveratrol antiviral effects, including regulation of important cellular pathways such as those involved in cell cycle, apoptosis and inflammatory responses [61]. In this regard, resveratrol inhibits the NF-κB pathway through several mechanisms. NF-κB is an important transcription factor, considered a fundamental regulator of the inflammatory cellular response, and it is also involved in cellular proliferation, transformation and tumor development [62,63,64]. Different stimuli, for example, those elicited during bacterial and viral infections, can activate NF-κB, and consequently its role for NF-κB in the innate immune response to infection has been largely described [65]. Resveratrol inhibits the activation of the NF-κB pathway by blocking TNF-α production in human monocyte and macrophage cell lines in vitro and in mouse skin in vivo (topical application of resveratrol 0.25–1 μmol for 4 h) by hampering phosphorylation of IκB by IKK, and as a result, hindering the translocation of NF-κB to the nucleus [66,67,68]. These results shed light on the antiviral effects mediated by resveratrol. In fact, although usually NF-κB activation has a protective role, some viruses rely on NF-κB activation for efficient replication [69], as is the case for influenza A virus [70] and HSV-1 [71]. Consequently, suppression of the NF-κB pathway seems to be one of the mechanisms through which resveratrol exerts its antiviral activity [54].
Further emphasizing its role as an anti-inflammatory molecule with potentially useful therapeutically implications, it was observed that resveratrol reduces the expression of several genes with proinflammatory functions, including TNF-α, CCL3, IL-1β in patients with diabetes and with coronary heart diseases [72]. Moreover, resveratrol has the potential to control the levels of inflammation in healthy subjects, since it reduces the expression of a number of other inflammatory and oxidative cytokines and stress markers such as p47phox, CRP, IL-6, JNK1, IKKβ, TLR4 [73,74]. In the emerging field of miRNAs as new signaling molecules, resveratrol seems to exert some anti-inflammatory effects in the monocytoid cell line THP-1, by upregulating miR-663, a miRNA which targets several genes involved in the immune response [75].
A large body of literature emphasizes the effect of resveratrol on enzyme activities or its binding to receptors, activating cellular pathways involved in apoptosis [76,77]. In this regard, it has been shown that upon entry via endocytosis, resveratrol-induced apoptosis follows its accumulation in lipid rafts triggering activation of MAPK and caspases [78]. With a different mechanism, resveratrol has been involved in p53-mediated apoptosis [79,80,81,82], exerted through increased p53-mediated transcriptional activity [83,84,85]. P53 is arguably the most important anticancer protein, considered the “guardian of the genome”, because of its crucial importance in preventing cellular transformation [86]. However, its stimulating effects are counteracted by activation of Sirt1, which has been reported to be one of the main targets of resveratrol [77]; indeed, the activation of Sirt1 may contribute to the beneficial effects of resveratrol in several diseases [87,88,89]. Sirt1 in turn interacts with and deacetylates p53, thus reducing p53-mediated functions [90,91]. Additional studies have highlighted the inhibitory effects of resveratrol on cyclo-oxygenases 1 and 2 (COX 1 and 2), two pro-inflammatory enzymes [92] and its effects on apoptosis [76,93,94]. The apoptosis or survival of viral-infected cells will ultimately depend on the preferential activation by resveratrol of Sirt1 or p53, and on resveratrol’s effects on the NF-κB pathway.
Another important cellular function influenced by resveratrol is related to cell cycle control. In this regard, by binding the αvβ3 integrin receptor, resveratrol hampers the functions of the IGF-1 receptor, resulting in reduced cells proliferation abilities [95]. A broad list of targets affected by resveratrol includes some cell cycle proteins with nuclear localization, phosphatases and cyclins [96,97]. All these data support the hypothesis that resveratrol is able to limit cellular proliferation. It therefore is not surprising that resveratrol has also been studied for its potential anticancer therapeutic properties.
Indeed, a number of in vitro data generated in different cancer cell lines, including stomach [98], colorectal [99], intestine [100], liver [101,102,103], kidney [104], blood [105,106,107,108], lung [109,110,111], ovaries [112,113], prostate [114,115], breast [116], glioma cells [117,118,119,120], head and neck tumors [121,122], bone [123], skin [124] and heart [125], as well as experiments in vivo in several animal models [19,126], have shown the anticancer mechanisms of resveratrol [127] and supported its use for cancer prevention due to its direct antitumor effects [128,129,130,131,132]. The route of administration varied from intragastric intubation to intraperitoneal injection, through diet or gavage. The amount also varied, from 10 mg/kg to 100 mg/kg daily, and the time of administration was from a few days to several weeks, indicating the absence of toxicity of resveratrol, but also the need to carefully determine the proper route and level of administration for an effective action [19]. In this regard, resveratrol intervenes in different stages of cellular transformation, affecting pathways usually dysregulated in cancer, including initiation, promotion to progression. In fact, resveratrol has been demonstrated to be involved in cell growth and division, inflammation, apoptosis, metastasis and angiogenesis [21,133]. Though in vivo and in vitro experiments have demonstrated generally positive outcomes, a certain variability was observed with regard to the route of administration, dose, tumor model and species. Regarding its use in human subjects, several data show promising results. In fact, a number of clinical trials have indicated the potential therapeutic and chemo preventive properties of resveratrol [21,131,134].
For example, administration of resveratrol resulted in 39% increase of the expression of cleaved caspase-3, a marker of apoptosis, in a phase I study in patients with hepatic metastases in malignant hepatic tissue, as compared with tissues from the patients treated with placebo [135], indicating a potential better response. In another study, repeated administration of Resveratrol in healthy volunteers decreased the levels of circulating IGF-I and IGFBP-3. Increased levels of these markers are associated with several types of cancer, since they have strong antiapoptotic and mitogenic properties and dysregulate cell differentiation, leading to increased neoplastic transformation and metastasis [136]. In an additional study investigating high levels of resveratrol consumption, 0.5 or 1.0 g of resveratrol administered daily before surgical resection resulted in 5% reduction of tumor cells proliferation in patients with colorectal cancer, supporting the use of resveratrol as a potential coadjuvant in specific anticancer treatments [137,138]. Finally, the effects of resveratrol on DNA methylation of cancer-related genes CCND-2, p16 and RASSF-1α were evaluated in women with high breast cancer risk. After 5–50 mg of resveratrol taken twice a day for 12 weeks, only RASSF-1 methylation was significantly reduced by resveratrol, while PGE2 (a lipid derived prostaglandin identified as a tumorigenic factor in many cancers [139]) was reduced in the nipple aspirate fluid (NAF) of the breast cancer patients that had undergone mammary ductoscopy [140]. Both these results indicated the potential beneficial effect of resveratrol in the treatment of breast cancer patients.
In fact, beside the previously described effects on p53-induction, several transcription factors implicated in cancer promotion or prevention have been shown to be affected by resveratrol, such as PPAR, PGC1α, NF-κB, Nrf2, c-Jun and CREB [141,142]. It is worth mentioning that low doses of resveratrol protected against chromosomal damage induced by four different genotoxins in both in vitro and in vivo experiments, further underscoring the efficacy of resveratrol as an agent that is able to prevent cell transformation [143]. Finally, treatment with resveratrol resulted in decreased levels of the specific oncogenic miRNAs targeting genes involved in the regulation of both tumor suppressors and effectors of the TGFβ-dependent signaling pathway [92,144,145,146,147].

2. Rapamycin

Rapamycin, also known as sirolimus, is a macrolide produced from streptomyces bacteria to inhibit fungal growth [148]. Rapamycin and its analogs inhibit the mammalian Target of Rapamycin (mTOR) complex 1 (mTORC1), an important molecule involved in cell proliferation. These drugs are currently approved by the FDA for the treatment of a number of conditions, as they have been demonstrated, both in vitro and in vivo, to slow aging, extend life span and prevent age-related diseases, including diabetic complications such as retinopathy [149].
Regarding the effect of rapamycin on the immune system, the molecule is considered an important immuno-modulator with immunosuppressive properties. mTOR (also known as FRAP1) is a major regulator of memory CD8+ T-cell differentiation. And contrary to what could be expected, administration of the immunosuppressive drug rapamycin results in immunostimulatory effects on the generation of memory CD8+ T cells, central players in the development of protective immunity. For this reason, eliciting an effective memory T-cell response is considered a major goal toward the development of functional vaccines against chronic infections and tumors. Consequently, several strategies have been employed for the improvement of vaccine strategies aimed at increasing the extent of the response relying on memory T-cells, and recently, several studies have focused on improving the functional qualities of the response (Table 2).
In this regard, it has been shown that rapamycin administered to mice intraperitoneally for about 30–40 days (75–600 μg/kg daily) following infection with acute lymphocytic choriomeningitis virus enhanced not only the amount, but also the quality/function of virus-specific CD8+ T cells [150]. Moreover, rapamycin (intra muscles for about 30–40 days (10–50 μg/kg daily) also increased memory T-cell responses in nonhuman primates after vaccination with modified Ankara virus, showing effectiveness during both phases of the T-cell response, namely expansion and contraction [150]: first, it increased the number of memory precursors, and second (during the effector to memory transition phase) it increased the differentiation of memory T-cell [150].
As a potential treatment against hepatitis B virus (HBV), rapamycin strongly inhibited the interaction between HBV large surface antigen (LHBs) and HBV entry receptor, sodium taurocholate cotransporting polypeptide (NTCP). This resulted in a strong reduction in hepatocyte infection with HBV with minimal cytotoxicity. The specific mechanism was identified in the direct interaction of rapamycin with NTCP, as demonstrated by surface plasmon analysis. Of note, rapamycin also prevented infection by hepatitis D virus, which also uses NTCP to enter cells [151].
Regarding Epstein-Barr virus (EBV), rapamycin inhibition of the mTOR pathway reduced total levels of BZLF1 transcripts, an immediate-early gene [152]. In addition, rapamycin alters the production of transcripts both in a cell-type, and in variant-type specific pattern, suggesting that the differential expression of these transcripts may control the replication cycle of EBV following infection [153].
Regarding HIV infection, rapamycin treatment of CD4 T cells in vitro reduces their CCR5 density levels (a coreceptor necessary for viral entry into the cells) and inhibits HIV-1 replication of CCR5-dependent virions (R5 strains). Not surprisingly, rapamycin improved the antiviral activity of T-20, an HIV-fusion inhibitor peptide used in therapy against R5 strains [154]. These data indicate that rapamycin may be used in clinical settings to increase the antiviral effect of T-20 against R5 strains, and, by extension, of other drugs targeting CCR5 [155,156,157]. Additional strategies have also been explored to exploit the combination of immune activation and the immunosuppressive properties of the mTOR inhibitor rapamycin in conjunction with T cell-activating agents in HIV-1 cure strategies [158]. Of note, rapamycin treatment did not impair cytotoxic T lymphocyte (CTL) recognition or the killing of HIV infected cells [158].
Recently, to evaluate its effect in the course of SARS-CoV-2 infection and the related cytokine storm, rapamycin (sirolimus) (6 mg on day 1 plus 2 mg daily for 13 days or until hospital discharge, whatever happens sooner) was used in clinical trials phase II in a randomized double blind placebo controlled study for the treatment of hospitalized patients with COVID-19. During the trial, several specific biomarkers were measured at day 3, 7 and 14; the study is supposed to conclude and post results soon (https://clinicaltrials.gov/ct2/show/NCT04341675).
The field of oncolytic viruses as treatment for cancer patients is currently expanding. Numerous clinical trials are ongoing, highlighting the vast clinical potential of this therapeutic tool. The current emphasis is on combining these viruses with molecules that upregulate viral replication, thus increasing oncolysis. Based on potential mechanisms by which mTOR inhibitors might enhance viral oncolysis, the antitumor effects of the combination of rapamycin and reovirus was tested in B16F10 cell, a murine model of malignant melanoma, providing evidence of synergistic antitumor cytotoxicity [159].

3. Metformin

Metformin is a natural compound derived from Galega officinalis. It is a widely used antihyperglycemic with a good safety profile and very few side-effects; additionally, it is inexpensive, and its usage has been associated with weight loss. Metformin is administered as an oral drug and is currently used to therapeutically treat patients affected by type-2 diabetes mellitus.
The mechanisms of action of metformin have been extensively studied [160,161,162]. Several cellular targets have been identified [163,164]; in particular, it inhibits the respiratory complex I of the electron transport chain [165,166], directly affecting reactions requiring ATP and indirectly acting on the activation of AMPK [161,164,165]. This, in turn, results in the inhibition of fatty acid synthesis and of the target of the rapamycin signaling network (mTOR) [167], causing reduced cellular energy consumption. An additional consequence of complex I inhibition is associated with increased accumulation of NADH compared to NAD+, affecting cellular biochemistry [168]. Indeed, metformin inhibits the protein NADH-ubiquinone oxidoreductase, which is localized on the mitochondrial membrane, thus activating AMPK and suppressing gluconeogenesis [169,170,171].
Metformin has also been shown to synergize with several anticancer drugs and help to reduce the chemo- and/or radio-resistance of different tumors. In addition, several data obtained both in vitro and in vivo suggest antiproliferative effects on tumor cells, and several molecular mechanisms have been described to account for these observations. In this regard, a number of clinical observations over the last few years indicate that metformin (used in doses between 0.5 and 1.5 mg/daily for at least six months) seems to reduce the risk of cancer development in diabetic patients and improve response to certain therapies and survival time in patients with certain types of cancers, such as non-small cell lung cancer [172,173,174,175], gastric cancer [176], colorectal cancer [177,178], prostate cancer [178,179], breast cancer [180,181] and pancreatic cancer [182,183]. These data, though still awaiting further validation, provide the basis for the use of metformin as an adjuvant therapy against cancer development and progression [184,185,186].
Recently, it was shown that metformin (1.7 mg/day for four months) caused important changes in the composition of the gut microbiota in diabetic patients, contributing to a better effect of the treatment [187]. In addition, in mice with endotoxemia-induced lung injury, metformin (400 mg/kg, intraperitoneal injection 30 min before LPS exposure) reduced levels of proinflammatory cytokines and improved survival [188]. These microbe-modulating properties of metformin prompted the study of several therapeutic areas of employment for metformin (Table 3).
Indeed, metformin showed efficacy as an antimicrobial agent against Trypanosomiasis cruzi, both in vitro and in mice (administered by gavage 400 mg/Kg/daily starting 20 days before infection and continuing for 71 days after infection) [189]. It also showed efficacy as an adjuvant in a retrospective analysis of diabetic patients with tuberculosis, and concomitantly showed that mice treated with 250 mg/kg/daily for three weeks had better outcome when infected with TB [190]. In vitro, the drug was active against Trichinella spiralis, Staphylococcus aureus and Pseudomonas aeruginosa, and there are indications of its potential beneficial role in sepsis, strongly suggesting that it could have a role as an agent or an adjuvant for the treatment of multiple infectious diseases [191].
When administered in combination with antiviral compounds such as interferon-α2b or Lamivudine (LMV), metformin enhanced their inhibitory effects, as measured by reduced Hepatitis B virus surface antigen (HBsAg) expression and HBV replication in vitro. An analysis of the mechanisms of action at a molecular level indicated that metformin works by partially hampering multiple HBV cis-acting elements. Based on these data, metformin could be potentially clinically helpful as an adjuvant to inhibit the HBsAg production in the treatment of HBV infection [192].
Infection with Hepatitis C virus (HCV) is associated with increased risk of hepatocellular carcinoma (HCC). On the other hand, treatments with metformin and statins show a protective effect. Following metformin and simvastatin treatment in vitro of human primary hepatocytes, a reduction was observed in mTOR and PTEN levels concomitant with an upregulation of levels of p62, LC3BII and Caspase 3, suggesting that this treatment could be useful in therapeutic prevention of HCV-related hepatocellular carcinoma [193].
Finally, metformin has been proposed as a potential coadjuvant treatment for COVID-19 patients [194,195,196]; this hypothesis is further supported by the fact that metformin can both reduce levels of IL-6 (important mediator of inflammation in COVID-19 patients) [197] and directly act on the pathways increasing cellular pH and subsequently interfering with the endocytic cycle, thus reducing viral replication [198,199].

4. Conclusions

Resveratrol, rapamycin and metformin are able to interfere with antiviral pathways and reduce viral replication and virus-related diseases, as demonstrated by experiments both in vitro and in vivo. This has prompted their use in several clinical settings. Additional studies aimed at better employing these compounds are needed, together with pharmacological strategies and dietary strategies to help strengthen their antiviral functions.

Author Contributions

All authors wrote sections of this manuscript and approved the submitted version. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Burns, J.; Yokota, T.; Ashihara, H.; Lean, M.E.J.; Crozier, A. Plant foods and herbal sources of resveratrol. J. Agric. Food Chem. 2002, 50, 3337–3340. [Google Scholar] [CrossRef] [PubMed]
  2. Salehi, B.; Mishra, A.P.; Nigam, M.; Sener, B.; Kilic, M.; Sharifi-Rad, M.; Fokou, P.V.T.; Martins, N.; Sharifi-Rad, J. Resveratrol: A Double-Edged Sword in Health Benefits. Biomedicines 2018, 6, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Walle, T.; Hsieh, F.; DeLegge, M.H.; Oatis, J.E.; Walle, U.K. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab. Dispos. 2004, 32, 1377–1382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Walle, T. Bioavailability of resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 9–15. [Google Scholar] [CrossRef]
  5. Pannu, N.; Bhatnagar, A. Resveratrol: From enhanced biosynthesis and bioavailability to multitargeting chronic diseases. Biomed. Pharmacother. 2019, 109, 2237–2251. [Google Scholar] [CrossRef]
  6. Gambini, J.; Inglés, M.; Lopez-Grueso, R.; Bonet-Costa, V.; Gimeno-Mallench, L.; Mas-Bargues, C. Properties of Resveratrol: In Vitro and In Vivo Studies about Metabolism, Bioavailability, and Biological Effects in Animal Models and Humans. Oxid. Med. Cell Longev. 2015, 2015, 837042. [Google Scholar] [CrossRef] [Green Version]
  7. Delmas, D.; Aires, V.; Limagne, E.; Dutartre, P.; Mazué, F.; Ghiringhelli, F.; Latruffe, N. Transport, stability, and biological activity of resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 48–59. [Google Scholar] [CrossRef]
  8. Boocock, D.; Patel, K.R.; Faust, G.E.; Normolle, D.P.; Marczylo, T.H.; Crowell, J.A.; Brenner, D.E.; Booth, T.D.; Gescher, A.; Steward, W.P. Quantitation of trans-resveratrol and detection of its metabolites in human plasma and urine by high performance liquid chromatography. J. Chromatogr. B 2007, 848, 182–187. [Google Scholar] [CrossRef] [Green Version]
  9. Baur, J.A.; Pearson, K.J.; Price, N.L.; Jamieson, H.A.; Lerin, C.; Kalra, A.; Prabhu, V.V.; Allard, J.S.; Lopez-Lluch, G.; Lewis, K.; et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature 2006, 444, 337–342. [Google Scholar] [CrossRef]
  10. Bhullar, K.S.; Hubbard, B.P. Lifespan and healthspan extension by resveratrol. Biochim. Biophys. Acta 2015, 1852, 1209–1218. [Google Scholar] [CrossRef] [Green Version]
  11. Pallauf, K.; Rimbach, G.; Rupp, P.M.; Chin, D.; Wolf, I.M.; Rimbacha, G.; Ruppa, P.; China, D. Resveratrol and Lifespan in Model Organisms. Curr. Med. Chem. 2016, 23, 4639–4680. [Google Scholar] [CrossRef]
  12. Galiniak, S.; Aebisher, D.; Bartusik-Aebisher, D. Health benefits of resveratrol administration. Acta Biochim. Pol. 2019, 66, 13–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Breuss, J.M.; Atanasov, A.G.; Uhrin, P. Resveratrol and Its Effects on the Vascular System. Int. J. Mol. Sci. 2019, 20, 1523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. He, S.; Yan, X. From resveratrol to its derivatives: New sources of natural antioxidant. Curr. Med. Chem. 2013, 20, 1005–1017. [Google Scholar] [PubMed]
  15. Truong, V.L.; Jun, M.; Jeong, W.S. Role of resveratrol in regulation of cellular defense systems against oxidative stress. Biofactors 2018, 44, 36–49. [Google Scholar] [CrossRef] [PubMed]
  16. Li, Y.R.; Li, S.; Lin, C.C. Effect of resveratrol and pterostilbene on aging and longevity. Biofactors 2018, 44, 69–82. [Google Scholar] [CrossRef] [PubMed]
  17. Varoni, E.M.; Lo Faro, A.F.; Sharifi-Rad, J.; Iriti, M. Anticancer Molecular Mechanisms of Resveratrol. Front. Nutr. 2016, 3, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Dybkowska, E.; Sadowska, A.; Świderski, F.; Rakowska, R.; Wysocka, K. The occurrence of resveratrol in foodstuffs and its potential for supporting cancer prevention and treatment. A review. Rocz. Panstw. Zakl. Hig. 2018, 69, 5–14. [Google Scholar] [PubMed]
  19. Carter, L.G.; D’Orazio, J.A.; Pearson, K.J. Resveratrol and cancer: Focus on in vivo evidence. Endocr. Relat. Cancer 2014, 21, R209–R225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Jiang, Z.; Chen, K.; Cheng, L.; Yan, B.; Qian, W.; Cao, J.; Liang, C.; Wu, E.; Ma, Q.; Yang, W. Resveratrol and cancer treatment: Updates. Ann. N. Y. Acad. Sci. 2017, 1403, 59–69. [Google Scholar] [CrossRef] [PubMed]
  21. Ko, J.-H.; Sethi, G.; Um, J.-Y.; Shanmugam, M.K.; Arfuso, F.; Kumar, A.P.; Bishayee, A.; Ahn, K.S. The Role of Resveratrol in Cancer Therapy. Int. J. Mol. Sci. 2017, 18, 2589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Berman, A.Y.; Motechin, R.A.; Wiesenfeld, M.Y.; Holz, M.K. The therapeutic potential of resveratrol: A review of clinical trials. NPJ Precis. Oncol. 2017, 1, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Saiko, P.; Akos, S.; Walter, J.; Thomas, S. Resveratrol and its analogs: Defense against cancer, coronary disease and neurodegenerative maladies or just a fad? Mutat. Res. 2008, 658, 68–94. [Google Scholar] [CrossRef] [PubMed]
  24. de la Lastra, C.A.; Villegas, I. Resveratrol as an anti-inflammatory and anti-aging agent: Mechanisms and clinical implications. Mol. Nutr. Food Res. 2005, 49, 405–430. [Google Scholar] [CrossRef]
  25. Springer, M.; Moco, M. Resveratrol and Its Human Metabolites-Effects on Metabolic Health and Obesity. Nutrients 2019, 11, 143. [Google Scholar] [CrossRef] [Green Version]
  26. Pourhanifeh, M.H.; Shafabakhsh, R.; Reiter, R.J.; Asemi, Z. The Effect of Resveratrol on Neurodegenerative Disorders: Possible Protective Actions Against Autophagy, Apoptosis, Inflammation and Oxidative Stress. Curr. Pharm. Des. 2019, 25, 2178–2191. [Google Scholar] [CrossRef]
  27. Howitz, K.T.; Bitterman, K.J.; Cohen, H.Y.; Lamming, D.W.; Lavu, S.; Wood, J.G.; Zipkin, R.E.; Chung, P.; Kisielewski, A.; Zhang, L.-L.; et al. Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature 2003, 425, 191–196. [Google Scholar] [CrossRef]
  28. Viswanathan, M.; Kim, S.K.; Berdichevsky, A.; Guarente, L. A Role for SIR-2.1 Regulation of ER Stress Response Genes in Determining C. elegans Life Span. Dev. Cell 2005, 9, 605–615. [Google Scholar] [CrossRef] [Green Version]
  29. Valenzano, D.R.; Terzibasi, E.; Genade, T.; Cattaneo, A.; Domenici, L. Resveratrol Prolongs Lifespan and Retards the Onset of Age-Related Markers in a Short-Lived Vertebrate. Curr. Biol. 2006, 16, 296–300. [Google Scholar] [CrossRef] [Green Version]
  30. Abba, Y.; Hassim, H.; Hamzah, H.; Noordin, M.M. Antiviral Activity of Resveratrol against Human and Animal Viruses. Adv. Virol. 2015, 2015, 184241. [Google Scholar] [CrossRef] [Green Version]
  31. Palamara, A.T.; Nencioni, L.; Aquilano, K.; De Chiara, G.; Hernandez, L.; Cozzolino, F.; Ciriolo, M.R.; Garaci, E. Inhibition of influenza A virus replication by resveratrol. J. Infect. Dis. 2005, 191, 1719–1729. [Google Scholar] [CrossRef] [PubMed]
  32. Cheng, K.; Wu, Z.; Gao, B.; Xu, J. Analysis of influence of baicalin joint resveratrol retention enema on the TNF-α, SIgA, IL-2, IFN-γ of rats with respiratory syncytial virus infection. Cell Biochem. Biophys. 2014, 70, 1305–1309. [Google Scholar] [CrossRef] [PubMed]
  33. Liu, T.; Zang, N.; Zhou, N.; Li, W.; Xie, X.; Deng, Y.; Ren, L.; Long, X.; Li, S.; Zhou, L.; et al. Resveratrol inhibits the TRIF-dependent pathway by upregulating sterile alpha and armadillo motif protein, contributing to anti-inflammatory effects after respiratory syncytial virus infection. J. Virol. 2014, 88, 4229–4236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Xie, X.-H.; Zang, N.; Li, S.; Wang, L.-J.; Deng, Y.; He, Y.; Yang, X.-Q.; Liu, E. Resveratrol Inhibits respiratory syncytial virus-induced IL-6 production, decreases viral replication, and downregulates TRIF expression in airway epithelial cells. Inflammation 2012, 35, 1392–1401. [Google Scholar] [CrossRef]
  35. Zang, N.; Li, S.; Li, W.; Xie, X.; Ren, L.; Long, X.; Xie, J.; Deng, Y.; Fu, Z.; Xu, F.; et al. Resveratrol suppresses persistent airway inflammation and hyperresponsivess might partially via nerve growth factor in respiratory syncytial virus-infected mice. Int. Immunopharmacol. 2015, 28, 121–128. [Google Scholar] [CrossRef]
  36. Zang, N.; Xie, X.; Deng, Y.; Wu, S.; Wang, L.; Peng, C.; Li, S.; Ni, K.; Luo, Y.; Liu, E. Resveratrol-mediated gamma interferon reduction prevents airway inflammation and airway hyperresponsiveness in respiratory syncytial virus-infected immunocompromised mice. J. Virol. 2011, 85, 13061–13068. [Google Scholar] [CrossRef] [Green Version]
  37. Filardo, S.; Di Pietro, M.; Mastromarino, P.; Sessa, R. Therapeutic potential of resveratrol against emerging respiratory viral infections. Pharmacol. Ther. 2020, 107613. [Google Scholar] [CrossRef]
  38. Docherty, J.J.; Sweet, T.J.; Bailey, E.; Faith, S.A.; Booth, T. Resveratrol inhibition of varicella-zoster virus replication in vitro. Antivir. Res. 2006, 72, 171–177. [Google Scholar] [CrossRef]
  39. Espinoza, J.L.; Takami, A.; Trung, L.Q.; Kato, S.; Nakao, S. Resveratrol prevents EBV transformation and inhibits the outgrowth of EBV-immortalized human B cells. PLoS ONE 2012, 7, e51306. [Google Scholar] [CrossRef] [Green Version]
  40. Yiu, C.-Y.; Chen, S.-Y.; Chang, L.-K.; Chiu, Y.-F.; Lin, T.-P. Inhibitory effects of resveratrol on the Epstein-Barr virus lytic cycle. Molecules 2010, 15, 7115–7124. [Google Scholar] [CrossRef] [Green Version]
  41. Chen, X.; Qiao, H.; Liu, T.; Yang, Z.; Xu, L.; Xu, Y.; Ge, H.M.; Tan, R.; Li, E. Inhibition of herpes simplex virus infection by oligomeric stilbenoids through ROS generation. Antivir. Res. 2012, 95, 30–36. [Google Scholar] [CrossRef] [PubMed]
  42. Docherty, J.; Fu, M.; Hah, J.; Sweet, T.; Faith, S.A.; Booth, T. Effect of resveratrol on herpes simplex virus vaginal infection in the mouse. Antivir. Res. 2005, 67, 155–162. [Google Scholar] [CrossRef] [PubMed]
  43. Docherty, J.J.; Fu, M.M.H.; Stiffler, B.S.; Limperos, R.J.; Pokabla, C.M.; DeLucia, A.L. Resveratrol inhibition of herpes simplex virus replication. Antivir. Res. 1999, 43, 145–155. [Google Scholar] [CrossRef]
  44. Docherty, J.J.; Smith, J.S.; Fu, M.M.; Stoner, T.; Booth, T. Effect of topically applied resveratrol on cutaneous herpes simplex virus infections in hairless mice. Antivir. Res. 2004, 61, 19–26. [Google Scholar] [CrossRef]
  45. Clouser, C.L.; Chauhan, J.; Bess, M.A.; Van Oploo, J.L.; Zhou, D.; Dimick-Gray, S.; Mansky, L.M.; Patterson, S.E. Anti-HIV-1 activity of resveratrol derivatives and synergistic inhibition of HIV-1 by the combination of resveratrol and decitabine. Bioorg. Med. Chem. Lett. 2012, 22, 6642–6646. [Google Scholar] [CrossRef] [Green Version]
  46. Heredia, A.; Davis, C.; Amin, M.N.; Le, N.M.; Wainberg, M.A.; Oliveira, M.; Deeks, S.G.; Wang, L.-X.; Redfield, R.R. Targeting host nucleotide biosynthesis with resveratrol inhibits emtricitabine-resistant HIV-1. AIDS 2014, 28, 317–323. [Google Scholar] [CrossRef] [Green Version]
  47. Chan, C.N.; Trinité, B.; Levy, D.N. Potent Inhibition of HIV-1 Replication in Resting CD4 T Cells by Resveratrol and Pterostilbene. Antimicrob. Agents Chemother. 2017, 61. [Google Scholar] [CrossRef] [Green Version]
  48. Zhang, L.; Yuanyuan, L.; Zhiwen, G.; Yuyue, W.; Yun, J.; Jing, S.; Xiaopeng, X.; Lirong, Z. Resveratrol inhibits enterovirus 71 replication and pro-inflammatory cytokine secretion in rhabdosarcoma cells through blocking IKKs/NF-κB signaling pathway. PLoS ONE 2015, 10, e0116879. [Google Scholar] [CrossRef] [Green Version]
  49. Du, N.; Li, X.; Bao, W.; Wang, B.; Xu, G.; Wang, F. Resveratrol-loaded nanoparticles inhibit enterovirus 71 replication through the oxidative stress-mediated ERS/autophagy pathway. Int. J. Mol. Med. 2019, 44, 737–749. [Google Scholar] [CrossRef]
  50. Nakamura, M.; Saito, H.; Ikeda, M.; Hokari, R.; Kato, N.; Hibi, T.; Miura, S. An antioxidant resveratrol significantly enhanced replication of hepatitis C virus. World J. Gastroenterol. 2010, 16, 184–192. [Google Scholar] [CrossRef]
  51. Long, X.; Li, S.; Xie, J.; Li, W.; Zang, N.; Ren, L.; Deng, Y.; Xie, X.; Wang, L.; Fu, Z.; et al. MMP-12-mediated by SARM-TRIF signaling pathway contributes to IFN-γ-independent airway inflammation and AHR post RSV infection in nude mice. Respir. Res. 2015, 16, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Kapadia, G.J.; Azuine, M.A.; Tokudab, H.; Takasakic, M.; Mukainakab, T.; Konoshimac, T.; Nishinob, H. Chemopreventive effect of resveratrol, sesamol, sesame oil and sunflower oil in the Epstein-Barr virus early antigen activation assay and the mouse skin two-stage carcinogenesis. Pharmacol. Res. 2002, 45, 499–505. [Google Scholar] [CrossRef] [PubMed]
  53. De Leo, A.; Arena, G.; Lacanna, E.; Oliviero, G.; Colavita, F.; Mattia, E. Resveratrol inhibits Epstein Barr Virus lytic cycle in Burkitt’s lymphoma cells by affecting multiple molecular targets. Antivir. Res. 2012, 96, 196–202. [Google Scholar] [CrossRef] [PubMed]
  54. Faith, S.A.; Sweet, T.J.; Bailey, E. Resveratrol suppresses nuclear factor-kappaB in herpes simplex virus infected cells. Antivir. Res. 2006, 72, 242–251. [Google Scholar] [CrossRef] [PubMed]
  55. Han, Y.S.; Quashie, P.K.; Mesplède, T. A resveratrol analog termed 3,3′,4,4′,5,5′-hexahydroxy-trans-stilbene is a potent HIV-1 inhibitor. J. Med. Virol. 2015, 87, 2054–2060. [Google Scholar] [CrossRef] [PubMed]
  56. Lin, S.C.; Ho, C.T.; Chou, W.H. Effective inhibition of MERS-CoV infection by resveratrol. BMC Infect. Dis. 2017, 17, 144. [Google Scholar] [CrossRef] [Green Version]
  57. Horne, J.R.; Vohl, M.C. Biological plausibility for interactions between dietary fat, resveratrol, ACE2, and SARS-CoV illness severity. Am. J. Physiol. Endocrinol. Metab. 2020, 318, E830–E833. [Google Scholar] [CrossRef] [Green Version]
  58. Marinella, M.A. Indomethacin and resveratrol as potential treatment adjuncts for SARS-CoV-2/COVID-19. Int. J. Clin. Pract. 2020, e13535. [Google Scholar] [CrossRef]
  59. Huang, C.; Wang, L.; Li, X.; Ren, L.; Zhao, J.; Hu, L.; Zhang, L. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
  60. AlGhatrif, M.; Cingolani, O.; Lakatta, E.G. The Dilemma of Coronavirus Disease 2019, Aging, and Cardiovascular Disease: Insights From Cardiovascular Aging Science. JAMA Cardiol. 2020, 5, 747–748. [Google Scholar] [CrossRef] [Green Version]
  61. Campagna, M.; Rivas, C. Antiviral activity of resveratrol. Biochem. Soc. Trans. 2010, 38, 50–53. [Google Scholar] [CrossRef] [PubMed]
  62. Li, Q.; Verma, I.M. NF-kappaB regulation in the immune system. Nat. Rev. Immunol. 2002, 2, 725–734. [Google Scholar] [CrossRef] [PubMed]
  63. Benedetti, F.; Curreli, S.; Krishnan, S.; Davinelli, S.; Cocchi, F.; Scapagnini, G.; Gallo, R.C.; Zella, D. Anti-inflammatory effects of H2S during acute bacterial infection: A review. J. Transl. Med. 2017, 15, 100. [Google Scholar] [CrossRef] [PubMed]
  64. Benedetti, F. Sulfur compounds block MCP-1 production by Mycoplasma fermentans-infected macrophages through NF-κB inhibition. J. Transl. Med. 2014, 12, 145. [Google Scholar] [CrossRef] [Green Version]
  65. Hiscott, J.; Kwon, H.; Génin, P. Hostile takeovers: Viral appropriation of the NF-kappaB pathway. J. Clin. Investig. 2001, 107, 143–151. [Google Scholar] [CrossRef]
  66. Holmes-McNary, M.; Baldwin, A.S., Jr. Chemopreventive properties of trans-resveratrol are associated with inhibition of activation of the IkappaB kinase. Cancer Res. 2000, 60, 3477–3483. [Google Scholar]
  67. Kundu, J.K.; Shin, Y.K.; Kim, S.H.; Surh, Y.J. Resveratrol inhibits phorbol ester-induced expression of COX-2 and activation of NF-kappaB in mouse skin by blocking IkappaB kinase activity. Carcinogenesis 2006, 27, 1465–1474. [Google Scholar] [CrossRef]
  68. Manna, S.K.; Mukhopadhyay, A.; Aggarwal, B.B. Resveratrol suppresses TNF-induced activation of nuclear transcription factors NF-kappa B, activator protein-1, and apoptosis: Potential role of reactive oxygen intermediates and lipid peroxidation. J. Immunol. 2000, 164, 6509–6519. [Google Scholar] [CrossRef] [Green Version]
  69. Santoro, M.G.; Rossi, A.; Amici, C. NF-kappaB and virus infection: Who controls whom. EMBO J. 2003, 22, 2552–2560. [Google Scholar] [CrossRef]
  70. Nimmerjahn, F.; Diana, D.; Ulrike, D.; Gerd, H.; Alexander, R.; Martin, S.; Louis, M.S.; Andreas, R.; Uta, B.; Georg, W.B.; et al. Active NF-kappaB signalling is a prerequisite for influenza virus infection. J. Gen. Virol. 2004, 85, 2347–2356. [Google Scholar] [CrossRef]
  71. Goodkin, M.L.; Ting, A.T.; Blaho, J.A. NF-kappaB is required for apoptosis prevention during herpes simplex virus type 1 infection. J. Virol. 2003, 77, 7261–7280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Tomé-Carneiro, J.; Gonzálvez, M.; Larrosa, M.; Yáñez-Gascón, M.J.; García-Almagro, F.J.; Ruiz-Ros, J.A.; Tomás-Barberán, F.A.; García-Conesa, M.T.; Espín, J.C. Grape resveratrol increases serum adiponectin and downregulates inflammatory genes in peripheral blood mononuclear cells: A triple-blind, placebo-controlled, one-year clinical trial in patients with stable coronary artery disease. Cardiovasc. Drugs Ther. 2013, 27, 37–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Rafe, T.; Shawon, P.A.; Salem, L.; Chowdhury, N.I.; Kabir, F.; Bin Zahur, S.M.; Akhter, R.; Noor, H.B.; Mohib, M.; Sagor, A.T.; et al. Preventive Role of Resveratrol Against Inflammatory Cytokines and Related Diseases. Curr. Pharm. Des. 2019, 25, 1345–1371. [Google Scholar] [CrossRef]
  74. Ghanim, H.; Sia, C.L.; Abuaysheh, S.; Korzeniewski, K.; Patnaik, P.; Marumganti, A.; Chaudhuri, A.; Dandona, P. An antiinflammatory and reactive oxygen species suppressive effects of an extract of Polygonum cuspidatum containing resveratrol. J. Clin. Endocrinol. Metab. 2010, 95, E1–E8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Tili, E.; Michaille, J.-J.; Adair, B.; Alder, H.; Limagne, E.; Taccioli, C.; Ferracin, M.; Delmas, D.; Latruffe, N.; Croce, C.M. Resveratrol decreases the levels of miR-155 by upregulating miR-663, a microRNA targeting JunB and JunD. Carcinogenesis 2010, 31, 1561–1566. [Google Scholar] [CrossRef] [PubMed]
  76. Lin, H.-Y.; Tang, H.-Y.; Davis, F.B.; Davis, P.J. Resveratrol and apoptosis. Ann. N. Y. Acad. Sci. 2011, 1215, 79–88. [Google Scholar] [CrossRef]
  77. Kulkarni, S.S.; Cantó, C. The molecular targets of resveratrol. Biochim. Biophys. Acta 2015, 1852, 1114–1123. [Google Scholar] [CrossRef] [Green Version]
  78. Colin, D.; Limagne, E.; Jeanningros, S.; Jacquel, A.; Lizard, G.; Athias, A.; Gambert, P.; Hichami, A.; Latruffe, N.; Solary, E.; et al. Endocytosis of resveratrol via lipid rafts and activation of downstream signaling pathways in cancer cells. Cancer Prev. Res. 2011, 4, 1095–1106. [Google Scholar] [CrossRef] [Green Version]
  79. Lin, H.Y.; Shih, A.I.; Faith, B.; Tang, H.Y.; Leon, J.M.; James, A.B.; Paul, J.D. Resveratrol induced serine phosphorylation of p53 causes apoptosis in a mutant p53 prostate cancer cell line. J. Urol. 2002, 168, 748–755. [Google Scholar] [CrossRef]
  80. Shih, A.; Faith, B.; Paul, J.D.; Lin, H.Y. Resveratrol induces apoptosis in thyroid cancer cell lines via a MAPK- and p53-dependent mechanism. J. Clin. Endocrinol. Metab. 2002, 87, 1223–1232. [Google Scholar] [CrossRef]
  81. Shih, A.; Zhang, S.; Cao, H.J.; Boswell, S.; Wu, Y.-H.; Tang, H.-Y.; Lennartz, M.R.; Davis, F.B.; Davis, P.J.; Lin, H.-Y. Inhibitory effect of epidermal growth factor on resveratrol-induced apoptosis in prostate cancer cells is mediated by protein kinase C-alpha. Mol. Cancer Ther. 2004, 3, 1355–1364. [Google Scholar] [PubMed]
  82. Zhang, S.; Cao, H.J.; Davis, F.B.; Tang, H.-Y.; Davis, P.J.; Lin, H.-Y. Oestrogen inhibits resveratrol-induced post-translational modification of p53 and apoptosis in breast cancer cells. Br. J. Cancer 2004, 91, 178–185. [Google Scholar] [CrossRef] [Green Version]
  83. Hsieh, T.C.; Juan, G.; Darzynkiewicz, Z.; Wu, J.M. Resveratrol increases nitric oxide synthase, induces accumulation of p53 and p21(WAF1/CIP1), and suppresses cultured bovine pulmonary artery endothelial cell proliferation by perturbing progression through S and G2. Cancer Res. 1999, 59, 2596–2601. [Google Scholar] [PubMed]
  84. Carbó, N.; Costelli, P.; Baccino, F.M.; López-Soriano, F.J.; Argilés, J.M. Resveratrol, a natural product present in wine, decreases tumour growth in a rat tumour model. Biochem. Biophys. Res. Commun. 1999, 254, 739–743. [Google Scholar] [CrossRef] [PubMed]
  85. Huang, C.; Ma, W.-Y.; Goranson, A.; Dong, Z. Resveratrol suppresses cell transformation and induces apoptosis through a p53-dependent pathway. Carcinogenesis 1999, 20, 237–242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Joerger, A.C.; Fersht, A.R. The p53 Pathway: Origins, Inactivation in Cancer, and Emerging Therapeutic Approaches. Annu. Rev. Biochem. 2016, 85, 375–404. [Google Scholar] [CrossRef]
  87. Price, N.L.; Gomes, A.P.; Ling, A.J.; Duarte, F.V.; Martin-Montalvo, A.; North, B.J.; Agarwal, B.; Ye, L.; Ramadori, G.; Teodoro, J.S.; et al. SIRT1 is required for AMPK activation and the beneficial effects of resveratrol on mitochondrial function. Cell Metab. 2012, 15, 675–690. [Google Scholar] [CrossRef] [Green Version]
  88. Maugeri, A.; Barchitta, M.; Mazzone, M.G.; Giuliano, F.; Basile, G.; Agodi, A. Resveratrol Modulates SIRT1 and DNMT Functions and Restores LINE-1 Methylation Levels in ARPE-19 Cells under Oxidative Stress and Inflammation. Int. J. Mol. Sci. 2018, 19, 2118. [Google Scholar] [CrossRef] [Green Version]
  89. Lee, S.-H.; Lee, J.-H.; Lee, H.-Y.; Min, A.K.-J. Sirtuin signaling in cellular senescence and aging. BMB Rep. 2019, 52, 24–34. [Google Scholar] [CrossRef] [Green Version]
  90. Lee, S.-H.; Lee, J.-H.; Lee, H.-Y.; Min, A.K.-J. Human SIR2 deacetylates p53 and antagonizes PML/p53-induced cellular senescence. EMBO J. 2002, 21, 2383–2396. [Google Scholar]
  91. Luo, J.; Nikolaev, A.Y.; Imai, S.-I.; Chen, D.; Su, F.; Shiloh, A.; Guarente, L.; Gu, W. Negative control of p53 by Sir2alpha promotes cell survival under stress. Cell 2001, 107, 137–148. [Google Scholar] [CrossRef] [Green Version]
  92. Latruffe, N.; Lançon, A.; Frazzi, R.; Aires, V.; Delmas, M.; Michaille, J.-J.; Djouadi, F.; Bastin, J.; Cherkaoui-Malki, M. Exploring new ways of regulation by resveratrol involving miRNAs, with emphasis on inflammation. Ann. N. Y. Acad. Sci. 2015, 1348, 97–106. [Google Scholar] [CrossRef]
  93. Bai, X.; Yao, L.; Ma, X.; Xu, X. Small Molecules as SIRT Modulators. Mini Rev. Med. Chem. 2018, 18, 1151–1157. [Google Scholar] [CrossRef] [PubMed]
  94. Pawlowska, E.; Szczepanska, J.; Szatkowska, M.; Blasiak, J. An Interplay between Senescence, Apoptosis and Autophagy in Glioblastoma Multiforme-Role in Pathogenesis and Therapeutic Perspective. Int. J. Mol. Sci. 2018, 19, 889. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Ho, Y.; Yang, Y.-C.; Chin, Y.-T.; Chou, S.-Y.; Chen, Y.-R.; Shih, Y.-J.; Whang-Peng, J.; Changou, C.A.; Liu, H.-L.; Lin, S.-J.; et al. Resveratrol inhibits human leiomyoma cell proliferation via crosstalk between integrin αvβ3 and IGF-1R. Food Chem. Toxicol. 2018, 120, 346–355. [Google Scholar] [CrossRef] [PubMed]
  96. Marel, A.K.; Gérard, L.; Izard, J.C.; Norbert, L.D. Inhibitory effects of trans-resveratrol analogs molecules on the proliferation and the cell cycle progression of human colon tumoral cells. Mol. Nutr. Food Res. 2008, 52, 538–548. [Google Scholar] [CrossRef] [Green Version]
  97. Colin, D.; Gimazane, A.; Lizard, G.; Izard, J.-C.; Solary, E.; Latruffe, N.; Delmas, D. Effects of resveratrol analogs on cell cycle progression, cell cycle associated proteins and 5fluoro-uracil sensitivity in human derived colon cancer cells. Int. J. Cancer 2009, 124, 2780–2788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Wu, X.; Xu, Y.; Zhu, B.; Liu, Q.; Yao, Q.; Zhao, G. Resveratrol induces apoptosis in SGC-7901 gastric cancer cells. Oncol. Lett. 2018, 16, 2949–2956. [Google Scholar] [CrossRef]
  99. Delmas, D.; Passilly-Degrace, P.; Jannin, B.; Malki, M.C.; Latruffe, N. Resveratrol, a chemopreventive agent, disrupts the cell cycle control of human SW480 colorectal tumor cells. Int. J. Mol. Med. 2002, 10, 193–199. [Google Scholar] [CrossRef]
  100. El-Readi, M.Z.; Eid, S.; Abdelghany, A.A.; Al-Amoudi, H.S.; Efferth, T.; Wink, M. Resveratrol mediated cancer cell apoptosis, and modulation of multidrug resistance proteins and metabolic enzymes. Phytomedicine 2019, 55, 269–281. [Google Scholar] [CrossRef]
  101. Liu, M.-H.; Lin, X.-L.; Li, J.; He, J.; Tan, T.-P.; Wu, S.-J.; Yu, S.; Chen, L.; Liu, J.; Tian, W.; et al. Resveratrol induces apoptosis through modulation of the Akt/FoxO3a/Bim pathway in HepG2 cells. Mol. Med. Rep. 2016, 13, 1689–1694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Delmas, D.; Jannin, B.; Malki, M.C.; Latruffe, N. Inhibitory effect of resveratrol on the proliferation of human and rat hepatic derived cell lines. Oncol. Rep. 2000, 7, 847–852. [Google Scholar] [CrossRef] [PubMed]
  103. Chai, R.; Fu, H.; Zheng, Z.; Liu, T.; Ji, S.; Li, G. Resveratrol inhibits proliferation and migration through SIRT1 mediated post-translational modification of PI3K/AKT signaling in hepatocellular carcinoma cells. Mol. Med. Rep. 2017, 16, 8037–8044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Kabel, A.M.; Atef, A.; Estfanous, R.S. Ameliorative potential of sitagliptin and/or resveratrol on experimentally-induced clear cell renal cell carcinoma. Biomed. Pharmacother. 2018, 97, 667–674. [Google Scholar] [CrossRef] [PubMed]
  105. Frazzi, R.; Valli, R.; Tamagnini, I.; Casali, B.; Latruffe, N.; Merli, F. Resveratrol-mediated apoptosis of hodgkin lymphoma cells involves SIRT1 inhibition and FOXO3a hyperacetylation. Int. J. Cancer 2013, 132, 1013–1021. [Google Scholar] [CrossRef]
  106. Frazzi, R.; Guardi, M. Cellular and Molecular Targets of Resveratrol on Lymphoma and Leukemia Cells. Molecules 2017, 22, 885. [Google Scholar] [CrossRef] [Green Version]
  107. Frazzi, R.; Tigano, M. The multiple mechanisms of cell death triggered by resveratrol in lymphoma and leukemia. Int. J. Mol. Sci. 2014, 15, 4977–4993. [Google Scholar] [CrossRef] [Green Version]
  108. Castello, L.; Tessitore, L. Resveratrol inhibits cell cycle progression in U937 cells. Oncol. Rep. 2005, 13, 133–137. [Google Scholar]
  109. Yousef, M.; Vlachogiannis, I.A.; Tsiani, E. Effects of Resveratrol against Lung Cancer: In Vitro and In Vivo Studies. Nutrients 2017, 9, 1231. [Google Scholar] [CrossRef] [Green Version]
  110. Wang, J.; Li, J.; Cao, N.; Li, Z.; Han, J.; Li, L. Resveratrol, an activator of SIRT1, induces protective autophagy in non-small-cell lung cancer via inhibiting Akt/mTOR and activating p38-MAPK. Onco Targets Ther. 2018, 11, 7777–7786. [Google Scholar] [CrossRef] [Green Version]
  111. Yuan, L.; Zhang, Y.; Xia, J.; Liu, B.; Zhang, Q.; Liu, J.; Luo, L.; Peng, Z.; Song, Z.; Zhu, R. Resveratrol induces cell cycle arrest via a p53-independent pathway in A549 cells. Mol. Med. Rep. 2015, 11, 2459–2464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Liu, Y.; Tong, L.; Luo, Y.; Li, X.; Chen, G.; Wang, Y. Resveratrol inhibits the proliferation and induces the apoptosis in ovarian cancer cells via inhibiting glycolysis and targeting AMPK/mTOR signaling pathway. J. Cell Biochem. 2018, 119, 6162–6172. [Google Scholar] [CrossRef] [PubMed]
  113. Zhang, Y.; Yang, S.; Yang, Y.; Liu, T. Resveratrol induces immunogenic cell death of human and murine ovarian carcinoma cells. Infect. Agent Cancer 2019, 14, 27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Aziz, M.H.; Nihal, M.; Fu, V.X.; Jarrard, D.F.; Ahmad, N. Resveratrol-caused apoptosis of human prostate carcinoma LNCaP cells is mediated via modulation of phosphatidylinositol 3′-kinase/Akt pathway and Bcl-2 family proteins. Mol. Cancer Ther. 2006, 5, 1335–1341. [Google Scholar] [CrossRef] [Green Version]
  115. Singh, S.K.; Saswati, B.; Edward, P.A.; James, W.L.; Rajesh, S. Resveratrol induces cell cycle arrest and apoptosis with docetaxel in prostate cancer cells via a p53/ p21WAF1/CIP1 and p27KIP1 pathway. Oncotarget 2017, 8, 17216–17228. [Google Scholar] [CrossRef] [Green Version]
  116. Sun, Y.; Zhou, Q.-M.; Lu, Y.-Y.; Zhang, H.; Chen, Q.; Zhao, M.; Shi-Bing, S. Resveratrol Inhibits the Migration and Metastasis of MDA-MB-231 Human Breast Cancer by Reversing TGF-β1-Induced Epithelial-Mesenchymal Transition. Molecules 2019, 24, 1131. [Google Scholar] [CrossRef] [Green Version]
  117. Pizarro, J.G.; Verdaguer, E.; Ancrenaz, V.; Junyent, F.; Sureda, F.; Pallàs, M.; Folch, J.; Camins, A. Resveratrol inhibits proliferation and promotes apoptosis of neuroblastoma cells: Role of sirtuin 1. Neurochem. Res. 2011, 36, 187–194. [Google Scholar] [CrossRef]
  118. Liu, L.; Zhang, Y.; Zhu, K.; Song, L.; Tao, M.; Huang, P.; Pan, Y. Resveratrol inhibits glioma cell growth via targeting LRIG1. J. BUON 2018, 23, 403–409. [Google Scholar]
  119. Cilibrasi, C.; Riva, G.; Romano, G.; Cadamuro, M.; Bazzoni, R.; Butta, V.; Paoletta, L.; Dalprà, L.; Strazzabosco, M.; Lavitrano, M.; et al. Resveratrol Impairs Glioma Stem Cells Proliferation and Motility by Modulating the Wnt Signaling Pathway. PLoS ONE 2017, 12, e0169854. [Google Scholar] [CrossRef] [Green Version]
  120. Song, Y.; Chen, Y.; Li, Y.; Lyu, X.; Cui, J.; Cheng, Y.; Zheng, T.; Zhao, L.; Zhao, G. Resveratrol Suppresses Epithelial-Mesenchymal Transition in GBM by Regulating Smad-Dependent Signaling. Biomed Res. Int. 2019, 2019, 1321973. [Google Scholar] [CrossRef] [Green Version]
  121. Zheng, T.; Feng, H.; Liu, L.; Peng, J.; Xiao, H.; Yu, T.; Zhou, Z.; Li, Y.; Zhang, Y.; Bai, X.; et al. Enhanced antiproliferative effect of resveratrol in head and neck squamous cell carcinoma using GE11 peptide conjugated liposome. Int. J. Mol. Med. 2019, 43, 1635–1642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Zhang, M.; Zhou, X.; Zhou, K. Resveratrol inhibits human nasopharyngeal carcinoma cell growth via blocking pAkt/p70S6K signaling pathways. Int. J. Mol. Med. 2013, 31, 621–627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Chen, G.; Xia, H.; Zhang, Z.-G.; Yu, H. Resveratrol in management of bone and spinal cancers. Nat. Prod. Res. 2019, 33, 516–526. [Google Scholar] [CrossRef] [PubMed]
  124. Zhao, H.; Han, L.; Jian, Y.; Ma, Y.; Yan, W.; Chen, X.; Xu, H.; Li, L. Resveratrol induces apoptosis in human melanoma cell through negatively regulating Erk/PKM2/Bcl-2 axis. Onco Targets Ther. 2018, 11, 8995–9006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Baarine, M.; Thandapilly, S.J.; Louis, X.L.; Mazué, F.; Yu, L.; Delmas, D.; Netticadan, T.; Lizard, G.; Latruffe, N. Pro-apoptotic versus anti-apoptotic properties of dietary resveratrol on tumoral and normal cardiac cells. Genes Nutr. 2011, 6, 161–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Jang, M.; Cai, L.; Udeani, G.O.; Slowing, K.V.; Thomas, C.F.; Beecher, C.W.W.; Fong, H.H.S.; Farnsworth, N.R.; Kinghorn, A.D.; Mehta, R.G.; et al. Cancer Chemopreventive Activity of Resveratrol, a Natural Product Derived from Grapes. Science 1997, 275, 218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Takashina, M.; Inoue, S.; Tomihara, K.; Tomita, K.; Hattori, K.; Zhao, Q.-L.; Suzuki, T.; Noguchi, M.; Ohashi, W.; Hattori, Y. Different effect of resveratrol to induction of apoptosis depending on the type of human cancer cells. Int. J. Oncol. 2017, 50, 787–797. [Google Scholar] [CrossRef]
  128. Trung, L.Q.; Espinoza, J.L.; Takami, A.; Nakao, S. Resveratrol induces cell cycle arrest and apoptosis in malignant NK cells via JAK2/STAT3 pathway inhibition. PLoS ONE 2013, 8, e55183. [Google Scholar] [CrossRef]
  129. Trung, L.Q.; Espinoza, J.L.; An, D.T.; Viet, N.H.; Shimoda, K.; Nakao, S. Resveratrol selectively induces apoptosis in malignant cells with the JAK2V617F mutation by inhibiting the JAK2 pathway. Mol. Nutr. Food Res. 2015, 59, 2143–2154. [Google Scholar] [CrossRef]
  130. Shukla, Y.; Singh, R. Resveratrol and cellular mechanisms of cancer prevention. Ann. N. Y. Acad. Sci. 2011, 1215, 1–8. [Google Scholar] [CrossRef]
  131. Vervandier-Fasseur, D.; Latruffe, N. The Potential Use of Resveratrol for Cancer Prevention. Molecules 2019, 24, 4506. [Google Scholar] [CrossRef] [Green Version]
  132. Rauf, A.; Imran, M.; Butt, M.S.; Nadeem, M.; Peters, D.G.; Mubarak, M.S. Resveratrol as an anti-cancer agent: A review. Crit. Rev. Food Sci. Nutr. 2018, 58, 1428–1447. [Google Scholar] [CrossRef]
  133. Rius, S.B.; Ametller, E.; Fuster, G.; Olivan, M.; Raab, V.; Argilés, J.M.; López-Soriano, F.J. Resveratrol, a natural diphenol, reduces metastatic growth in an experimental cancer model. Cancer Lett. 2007, 245, 144–148. [Google Scholar]
  134. Singh, A.P.; Singh, R.; Verma, S.S.; Rai, V.; Kaschula, C.H.; Maiti, P.; Gupta, S.C. Health benefits of resveratrol: Evidence from clinical studies. Med. Res. Rev. 2019, 39, 1851–1891. [Google Scholar] [CrossRef] [PubMed]
  135. Howells, L.M.; Berry, D.P.; Elliott, P.J.; Jacobson, E.W.; Hoffmann, E.; Hegarty, B.; Brown, K.; Steward, W.P.; Gescher, A.J. Phase I randomized, double-blind pilot study of micronized resveratrol (SRT501) in patients with hepatic metastases--safety, pharmacokinetics, and pharmacodynamics. Cancer Prev. Res. 2011, 4, 1419–1425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Brown, V.A.; Patel, K.R.; Viskaduraki, M.; Crowell, J.A.; Perloff, M.; Booth, T.D.; Vasilinin, G.; Sen, A.; Schinas, A.M.; Piccirilli, G.; et al. Repeat dose study of the cancer chemopreventive agent resveratrol in healthy volunteers: Safety, pharmacokinetics, and effect on the insulin-like growth factor axis. Cancer Res. 2010, 70, 9003–9011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Patel, K.R.; Brown, V.A.; Jones, D.J.L.; Britton, R.G.; Hemingway, D.; Miller, A.S.; West, K.P.; Booth, T.D.; Perloff, M.; Crowell, J.A.; et al. Clinical Pharmacology of Resveratrol and Its Metabolites in Colorectal Cancer Patients. Cancer Res. 2010, 70, 7392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Honari, M.; Shafabakhsh, R.; Reiter, R.J.; Mirzaei, H.; Asemi, Z. Resveratrol is a promising agent for colorectal cancer prevention and treatment: Focus on molecular mechanisms. Cancer Cell Int. 2019, 19, 180. [Google Scholar] [CrossRef] [Green Version]
  139. Ke, J.; Yang, Y.; Che, Q.; Jiang, F.; Wang, H.; Chen, Z.; Zhu, M.; Tong, H.; Zhang, H.; Yan, X.; et al. Prostaglandin E2 (PGE2) promotes proliferation and invasion by enhancing SUMO-1 activity via EP4 receptor in endometrial cancer. Tumour Biol. 2016, 37, 12203–12211. [Google Scholar] [CrossRef] [Green Version]
  140. Zhu, W.; Qin, W.; Zhang, K.; Rottinghaus, G.E.; Chen, Y.-C.; Kliethermes, B.; Sauter, E.R. Trans-resveratrol alters mammary promoter hypermethylation in women at increased risk for breast cancer. Nutr. Cancer 2012, 64, 393–400. [Google Scholar] [CrossRef] [Green Version]
  141. Delmas, D.; Lancon, A.; Colin, D.; Jannin, B.; Latruffe, N. Resveratrol as a chemopreventive agent: A promising molecule for fighting cancer. Curr. Drug Targets 2006, 7, 423–442. [Google Scholar] [CrossRef]
  142. Davinelli, S.; Sapere, N.; Visentin, M.; Zella, D.; Scapagnini, G. Enhancement of mitochondrial biogenesis with polyphenols: Combined effects of resveratrol and equol in human endothelial cells. Immun. Ageing 2013, 10, 1–5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Abraham, S.K.; Khandelwal, N.; Hintzsche, H.; Stopper, H. Antigenotoxic effects of resveratrol: Assessment of in vitro and in vivo response. Mutagenesis 2016, 31, 27–33. [Google Scholar] [PubMed] [Green Version]
  144. Tili, E.; Michaille, J.-J.; Alder, H.; Volinia, S.; Delmas, D.; Latruffe, N.; Croce, C.M. Resveratrol modulates the levels of microRNAs targeting genes encoding tumor-suppressors and effectors of TGFβ signaling pathway in SW480 cells. Biochem. Pharmacol. 2010, 80, 2057–2065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Alghetaa, H.; Mohammed, A.; Sultan, M.; Busbee, P.; Murphy, A.; Chatterjee, S.; Nagarkatti, M.; Nagarkatti, P. Resveratrol protects mice against SEB-induced acute lung injury and mortality by miR-193a modulation that targets TGF-β signalling. J. Cell Mol. Med. 2018, 22, 2644–2655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Zhang, Y.; Lu, Y.; Ong’Achwa, M.J.; Ge, L.; Qian, Y.; Chen, L.; Machuki, J.O.; Li, F.; Wei, H.; Zhang, C.; et al. Resveratrol Inhibits the TGF-β1-Induced Proliferation of Cardiac Fibroblasts and Collagen Secretion by Downregulating miR-17 in Rat. Biomed Res. Int. 2018, 2018, 8730593. [Google Scholar] [CrossRef] [Green Version]
  147. McCubrey, J.A.; Lertpiriyapong, K.; Steelman, L.S.; Abrams, S.L.; Yang, L.V.; Murata, R.M.; Rosalen, P.L.; Scalisi, A.; Neri, L.M.; Cocco, L.; et al. Effects of resveratrol, curcumin, berberine and other nutraceuticals on aging, cancer development, cancer stem cells and microRNAs. Aging 2017, 9, 1477–1536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Sehgal, S.; Baker, A.; Vezina, C. Rapamycin (AY-22, 989), A New Antifungal Antibiotic. J. Antibiot. 1975, 28, 727–732. [Google Scholar] [CrossRef] [Green Version]
  149. Blagosklonny, M.V. Fasting and rapamycin: Diabetes versus benevolent glucose intolerance. Cell Death Dis. 2019, 10, 607. [Google Scholar] [CrossRef] [Green Version]
  150. Araki, K.; Turner, A.P.; Shaffer, V.O.; Gangappa, S.; Keller, S.A.; Bachmann, M.F.; Larsen, C.P.; Ahmed, R. mTOR regulates memory CD8 T-cell differentiation. Nature 2009, 460, 108–112. [Google Scholar] [CrossRef] [Green Version]
  151. Saso, W.; Tsukuda, S.; Ohashi, H.; Fukano, K.; Morishita, R.; Matsunaga, S.; Ohki, M.; Ryo, A.; Park, S.-Y.; Suzuki, R.; et al. A new strategy to identify hepatitis B virus entry inhibitors by AlphaScreen technology targeting the envelope-receptor interaction. Biochem. Biophys. Res. Commun. 2018, 501, 374–379. [Google Scholar] [CrossRef] [PubMed]
  152. Adamson, A.L.; Le, B.T.; Siedenburg, B.D. Inhibition of mTORC1 inhibits lytic replication of Epstein-Barr virus in a cell-type specific manner. Virol. J. 2014, 11, 110. [Google Scholar] [CrossRef] [PubMed]
  153. Needham, J.; Adamson, A.L. BZLF1 transcript variants in Epstein-Barr virus-positive epithelial cell lines. Virus Genes 2019, 55, 779–785. [Google Scholar] [CrossRef] [PubMed]
  154. Heredia, A.; Gilliam, B.; Latinovic, O.; Le, N.; Bamba, D.; DeVico, A.; Melikyan, G.B.; Gallo, R.C.; Redfield, R.R. Rapamycin reduces CCR5 density levels on CD4 T cells, and this effect results in potentiation of enfuvirtide (T-20) against R5 strains of human immunodeficiency virus type 1 in vitro. Antimicrob. Agents Chemother. 2007, 51, 2489–2496. [Google Scholar] [CrossRef] [Green Version]
  155. Heredia, A.; Latinovic, O.; Gallo, R.C.; Melikyan, G.; Reitz, M.; Le, N.; Redfield, R.R. Reduction of CCR5 with low-dose rapamycin enhances the antiviral activity of vicriviroc against both sensitive and drug-resistant HIV-1. Proc. Natl. Acad. Sci. USA 2008, 105, 20476–20481. [Google Scholar] [CrossRef] [Green Version]
  156. Latinovic, O.S.; Neal, L.M.; Tagaya, Y.; Heredia, A.; Medina-Moreno, M.S.; Zapata, J.C.; Reitz, M.; Bryant, J.; Redfield, R.R. Suppression of Active HIV-1 Infection in CD34(+) Hematopoietic Humanized NSG Mice by a Combination of Combined Antiretroviral Therapy and CCR5 Targeting Drugs. AIDS Res. Hum. Retrovir. 2019, 35, 718–728. [Google Scholar] [CrossRef]
  157. Weichseldorfer, M.; Yvonne, A.; Alonso, H.; Yutaka, T.; Francesca, B.; Davide, Z.; Marvin, R.; Fabio, R.; Olga, S.L. Anti-HIV Activity of Standard cART in Primary Cells Is Intensified by CCR5-Targeting Drugs. AIDS Res. Hum. Retrovir. 2020, 36, 835–841. [Google Scholar] [CrossRef]
  158. Martin, A.R.; Pollack, R.A.; Capoferri, A.A.; Ambinder, R.F.; Durand, C.M.; Siliciano, R.F. Rapamycin-mediated mTOR inhibition uncouples HIV-1 latency reversal from cytokine-associated toxicity. J. Clin. Investig. 2017, 127, 651–656. [Google Scholar] [CrossRef] [Green Version]
  159. Comins, C.; Simpson, G.R.; Rogers, W.; Relph, K.; Harrington, K.; A Melcher, A.; Roulstone, V.; Kyula, J.; Pandha, H. Synergistic antitumour effects of rapamycin and oncolytic reovirus. Cancer Gene Ther. 2018, 25, 148–160. [Google Scholar] [CrossRef]
  160. Foretz, M.; Guigas, B.; Bertrand, L.; Pollak, M.; Viollet, B. Metformin: From mechanisms of action to therapies. Cell Metab. 2014, 20, 953–966. [Google Scholar] [CrossRef] [Green Version]
  161. Santos, S.; Marín, A.; Serra-Batlles, J.; De La Rosa, D.; Solanes, I.; Pomares, X.; López-Sánchez, M.; Muñoz-Esquerre, M.; Miravitlles, M. Treatment of patients with COPD and recurrent exacerbations: The role of infection and inflammation. Int. J. Chron. Obstruct. Pulmon. Dis. 2016, 11, 515–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Pryor, R.; Cabreiro, F. Repurposing metformin: An old drug with new tricks in its binding pockets. Biochem. J. 2015, 471, 307–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Kalender, A.; Selvaraj, A.; Kim, S.Y.; Gulati, P.; Brûlé, S.; Viollet, B.; Kemp, B.E.; Bardeesy, N.; Dennis, P.; Schlager, J.J.; et al. Metformin, independent of AMPK, inhibits mTORC1 in a rag GTPase-dependent manner. Cell Metab. 2010, 11, 390–401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Madiraju, A.K.; Erion, D.M.; Rahimi, Y.; Zhang, X.-M.; Braddock, D.T.; Albright, R.A.; Prigaro, B.J.; Wood, J.L.; Bhanot, S.; Macdonald, M.J.; et al. Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase. Nature 2014, 510, 542–546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Bridges, H.R.; Sirviö, V.A.; Agip, A.-N.A.; Hirst, J. Molecular features of biguanides required for targeting of mitochondrial respiratory complex I and activation of AMP-kinase. BMC Biol. 2016, 14, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Bridges, H.R.; Jones, A.J.Y.; Pollak, M.N.; Hirst, J. Effects of metformin and other biguanides on oxidative phosphorylation in mitochondria. Biochem. J. 2014, 462, 475–487. [Google Scholar] [CrossRef] [Green Version]
  167. Howell, J.J.; Hellberg, K.; Turner, M.; Talbott, G.; Kolar, M.J.; Ross, D.V.; Hoxhaj, G.; Saghatelian, A.; Shaw, R.J.; Manning, B.D. Metformin inhibits hepatic mTORC1 signaling via dose-dependent mechanisms involving AMPK and the TSC complex. Cell Metab. 2017, 25, 463–471. [Google Scholar] [CrossRef] [Green Version]
  168. Gui, D.Y.; Sullivan, L.B.; Luengo, A.; Hosios, A.M.; Bush, L.N.; Gitego, N.; Davidson, S.M.; Freinkman, E.; Thomas, C.J.; Heiden, M.G.V. Environment dictates dependence on mitochondrial complex I for NAD+ and aspartate production and determines cancer cell sensitivity to metformin. Cell Metab. 2016, 24, 716–727. [Google Scholar] [CrossRef] [Green Version]
  169. Shaw, R.J.; Lamia, K.A.; Vasquez, D.; Koo, S.-H.; Bardeesy, N.; Depinho, R.A.; Montminy, M.; Cantley, L.C. The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin. Science 2005, 310, 1642–1646. [Google Scholar] [CrossRef] [Green Version]
  170. Zhou, G.; Myers, R.; Li, Y.; Chen, X.; Shen, J.; Fenyk-Melody, M.; Wu, J.; Ventre, T.D.; Fujii, N.; Musi, N. Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Investig. 2001, 108, 1167–1174. [Google Scholar] [CrossRef]
  171. Rena, G.; Hardie, D.G.; Pearson, E.R. The mechanisms of action of metformin. Diabetologia 2017, 60, 1577–1585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Arrieta, O.; Varela-Santoyo, E.; Soto-Perez-De-Celis, E.; Sánchez-Reyes, R.; De La Torre-Vallejo, M.; Muñiz-Hernández, S.; Cardona, A.F. Metformin use and its effect on survival in diabetic patients with advanced non-small cell lung cancer. BMC Cancer 2016, 16, 633. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Sayed, R.; Saad, A.S.; El Wakeel, L.; Elkholy, E.; Badary, O. Metformin Addition to Chemotherapy in Stage IV Non-Small Cell Lung Cancer: An Open Label Randomized Controlled Study. Asian Pac. J. Cancer Prev. 2015, 16, 6621–6626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Morgillo, F.; Fasano, M.; Della Corte, C.M.; Sasso, F.C.; Papaccio, F.; Viscardi, G.; Esposito, G.; Di Liello, R.; Normanno, N.; Capuano, A.; et al. Results of the safety run-in part of the METAL (METformin in Advanced Lung cancer) study: A multicentre, open-label phase I-II study of metformin with erlotinib in second-line therapy of patients with stage IV non-small-cell lung cancer. ESMO Open 2017, 2, e000132. [Google Scholar] [CrossRef] [Green Version]
  175. Wan, G.; Yu, X.; Chen, P.; Wang, X.; Pan, D.; Wang, X.; Li, L.; Cai, X.; Cao, F. Metformin therapy associated with survival benefit in lung cancer patients with diabetes. Oncotarget 2016, 7, 35437–35445. [Google Scholar] [CrossRef] [Green Version]
  176. Tseng, C.-H. Metformin reduces gastric cancer risk in patients with type 2 diabetes mellitus. Aging 2016, 8, 1636–1649. [Google Scholar] [CrossRef] [Green Version]
  177. Park, J.W.; Lee, J.H.; Park, Y.; Park, S.J.; Cheon, J.H.; Kim, W.H.; Kim, T.I. Sex-dependent Difference in the effect of metformin on colorectal cancer-specific mortality of Diabetic colorectal cancer patients. World J. Gastroenterol. 2017, 23, 5196. [Google Scholar] [CrossRef]
  178. Coyle, C.; Cafferty, F.H.; Vale, C.; Langley, R.E. Metformin as an adjuvant treatment for cancer: A systematic review and meta-analysis. Ann. Oncol. 2016, 27, 2184–2195. [Google Scholar] [CrossRef]
  179. Haring, A.; Murtola, T.J.; Talala, K.; Taari, K.; Tammela, T.L.J.; Auvinen, A. Antidiabetic drug use and prostate cancer risk in the Finnish Randomized Study of Screening for Prostate Cancer. Scand. J. Urol. 2017, 51, 5–12. [Google Scholar] [CrossRef]
  180. He, X.; Esteva, F.J.; Ensor, J.; Hortobagyi, G.N.; Lee, M.-H.; Yeung, S.-C.J. Metformin and thiazolidinediones are associated with improved breast cancer-specific survival of diabetic women with HER2+ breast cancer. Ann. Oncol. 2012, 23, 1771–1780. [Google Scholar] [CrossRef]
  181. Lega, I.C.; Fung, K.; Austin, P.; Lipscombe, L. Metformin and breast cancer stage at diagnosis: A population-based study. Curr. Oncol. 2017, 24, e85–e91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Chen, K.; Qian, W.; Jiang, Z.; Cheng, L.; Liang, C.; Sun, L.; Zhou, C.; Gao, L.; Lei, M.; Yan, B.; et al. Metformin suppresses cancer initiation and progression in genetic mouse models of pancreatic cancer. Mol. Cancer 2017, 16, 131. [Google Scholar] [CrossRef] [PubMed]
  183. Li, X.; Li, T.; Liu, Z.; Gou, S.; Wang, C. The effect of metformin on survival of patients with pancreatic cancer: A meta-analysis. Sci. Rep. 2017, 7, 5825. [Google Scholar] [CrossRef] [PubMed]
  184. Christodoulou, M.I.; Scorilas, A. Metformin and Anti-Cancer Therapeutics: Hopes for a More Enhanced Armamentarium Against Human Neoplasias? Curr. Med. Chem. 2017, 24, 14–56. [Google Scholar] [CrossRef]
  185. Mallik, R.; Chowdhury, T.A. Metformin in cancer. Diabetes Res. Clin. Pract. 2018, 143, 409–419. [Google Scholar] [CrossRef]
  186. Chen, K.; Li, Y.; Guo, Z.; Zeng, Y.; Zhang, W.; Wang, H. Metformin: Current clinical applications in nondiabetic patients with cancer. Aging 2020, 12, 3993–4009. [Google Scholar] [CrossRef]
  187. Wu, H.; Esteve, E.; Tremaroli, V.; Khan, M.T.; Caesar, R.; Mannerås-Holm, L.; Ståhlman, M.; Olsson, L.M.; Serino, M.; Planas-Fèlix, M.; et al. Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug. Nature Med. 2017, 23, 850–858. [Google Scholar] [CrossRef]
  188. Wu, K.; Tian, R.; Huang, J.; Yang, Y.; Dai, J.; Jiang, R.; Zhang, L. Metformin alleviated endotoxemia-induced acute lung injury via restoring AMPK-dependent suppression of mTOR. Chemico-Biol. Interact. 2018, 291, 1–6. [Google Scholar] [CrossRef]
  189. Brima, W.; Daniel, J.; Syed, F.; Michelle, B.; Mohammad, M.; Joanna, S.; Vanessa, A.; Dazhi, Z.; Irwin, K.; Jeffrey, E.; et al. The brighter (and evolutionarily older) face of the metabolic syndrome: Evidence from Trypanosoma cruzi infection in CD-1 mice. Diabetes Metab. Res. Rev. 2015, 31, 346–359. [Google Scholar] [CrossRef] [Green Version]
  190. Singhal, A.; Jie, L.; Kumar, P.; Hong, G.S.; Leow, M.K.-S.; Paleja, B.; Tsenova, L.; Kurepina, N.; Chen, J.; Zolezzi, F.; et al. Metformin as adjunct antituberculosis therapy. Sci. Transl. Med. 2014, 6, 263ra159. [Google Scholar] [CrossRef]
  191. Malik, F.; Syed, F.M.; Ali, H.; Patel, P. Is metformin poised for a second career as an antimicrobial? Diabetes Metab. Res. Rev. 2018, 34, e2975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Xun, Y.-H.; Zhang, J.; Pan, Q.-C.; Mao, R.-C.; Qin, Y.-L.; Liu, H.-Y.; Yu, Y.-S.; Tang, Z.-H.; Lu, M.-J.; Zang, G.-Q. Metformin inhibits hepatitis B virus protein production and replication in human hepatoma cells. J. Viral Hepat. 2014, 21, 597–603. [Google Scholar] [CrossRef] [PubMed]
  193. Del Campo, J.; García-Valdecasas, M.; Gil-Gómez, A.; Rojas, Á.; Gallego, P.; Ampuero, J.; Gallego-Durán, R.; Pastor, H.; Grande, L.; Padillo, F.J.; et al. Simvastatin and metformin inhibit cell growth in hepatitis C virus infected cells via mTOR increasing PTEN and autophagy. PLoS ONE 2018, 13, e0191805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. El-Arabey, A.A.; Abdalla, M. Metformin and COVID-19: A novel deal of an old drug. J. Med. Virol. 2020. [Google Scholar] [CrossRef] [PubMed]
  195. Esam, Z. A proposed mechanism for the possible therapeutic potential of Metformin in COVID-19. Diabetes Res. Clin. Pract. 2020, 167, 108282. [Google Scholar] [CrossRef] [PubMed]
  196. Menendez, J.A. Metformin and SARS-CoV-2: Mechanistic lessons on air pollution to weather the cytokine/thrombotic storm in COVID-19. Aging 2020, 12, 8760–8765. [Google Scholar] [CrossRef] [PubMed]
  197. Blanco-Melo, D.; Payant, B.E.; Liu, W.C.; Hoagland, D.; Oishi, K.; Panis, M. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell 2020, 181, 1036–1045.e9. [Google Scholar] [CrossRef]
  198. Kim, J.; You, Y.-J. Regulation of organelle function by metformin. IUBMB Life 2017, 69, 459–469. [Google Scholar] [CrossRef] [Green Version]
  199. Zhang, C.-S.; Li, M.; Ma, T.; Zong, Y.; Cui, J.; Feng, J.-W.; Wu, Y.-Q.; Lin, S.-Y.; Lin, S.-C. Metformin Activates AMPK through the Lysosomal Pathway. Cell Metab. 2016, 24, 521–522. [Google Scholar] [CrossRef] [Green Version]
Table 1. Antiviral effects of resveratrol.
Table 1. Antiviral effects of resveratrol.
VirusAntiviral Effects
Influenza VirusBlock of nuclear-cytoplasmic translocation in decreased expression [31].
Respiratory syncytial virus (RPSV)Reduced inflammation and levels IFN-γ and TLR3; inhibition of TRIF signaling, induction of M2R [35,36]; decreased production of IL-6 and TBK1 [34]; increased expression of SARM and decreased expression of MMP-12 and TRIF leading to decreased IFN-γ expression and AHR [33,51]; reduced levels of NGF [36]; increased levels of TNF-α, IFN-γ, and IL-2 in infected mice [32].
Varicella Zoster virusDecreased synthesis of IE 62 [38].
Epstein-Barr virus (EBV)Inhibition of EBV early antigen and reduced papilloma production in mouse [52]; inhibition of EBV lytic cycle resulting in reduced production of viral particles [40]; inhibition of protein synthesis, reduction in ROS production, and inhibition of transcription factors NF-κB and AP1 [53]; prevention of EBV-mediated transformation of human B-cells [39].
Herpes simplex virus (HSV-1 and HSV-2)Decreased production of early viral protein ICP-4 and reduced production of viral particles; prevention of virus reactivation in latently infected neuron cells [43]; suppression of the development of cutaneous lesions in abraded skin infected with HSV-1 [44]; prevention of the development of vaginal lesions in mice infected with HSV-2 and HSV-1, with reduced mortality rate [42]; inhibition of the expression of immediate-early, early, and late HSV genes and viral DNA synthesis [41,54].
Human immunodeficiency virus (HIV)Resveratrol, decitabine and 15 other derivatives of resveratrol were potent antiviral drugs [45]: inhibition of DNA synthesis [46]; block of HIV-1 infection in resting CD4 T cells; 3,3′,4,4′,5,5′-hexahydroxy-trans-stilbene (M8) showed potent anti-HIV activity [55].
Enterovirus 71 (EV 71)Inhibition of viral protein 1 (VP1) synthesis and phosphorylation of proinflammatory cytokines in Rhabdomyosarcoma cell line [48].
MERS-CoVInhibition of MERS-CoV infection; downregulation of apoptosis induced by MERS-CoV in vitro [56].
SARS-CoV-2Upregulation of ACE-2 [57]; decreased high levels of circulating cytokines such as IL-6 and TNF-α, upregulated following SARS-CoV-2 infection [58].
Table 2. Antiviral effects of rapamycin on several viruses.
Table 2. Antiviral effects of rapamycin on several viruses.
VirusAntiviral Effects
Acute lymphocytic choriomeningitis virusEnhanced amount and quantity/function of virus specific CD8+ T cells [150].
Hepatitis B virus (HBV)Inhibition of the interaction between LHBs and NCTP and consequent infection reduction [151].
Epstein Bar Virus (EBV)Reduced levels of BZLF1 transcripts [152]; altered production of viral transcripts [153].
Human immunodeficiency virus (HIV)Reduced CCR5 levels and improvement of antiviral compound T20 [154,155,156,157].
Table 3. Antibacterial and antiviral effects of metformin.
Table 3. Antibacterial and antiviral effects of metformin.
Bacterium/VirusEffects
T. cruzi, T. spiralis, S. aureus, P. aeruginosaAntibacterial [189,190,191]
Hepatitis B Virus (HBV)Reduced HBsAg expression and viral replication [192]
Hepatitis C Virus (HCV)In combination with simvastatin, it was observed reduction of mTOR and PTEN levels, and upregulation of p62, LC3BII and caspase 3 in human primary hepatocytes in vitro [193]
SARS-CoV-2Reduction of IL-6 levels, increase of cellular pH levels and reduction of viral replication [194,195,196,197,198,199]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Benedetti, F.; Sorrenti, V.; Buriani, A.; Fortinguerra, S.; Scapagnini, G.; Zella, D. Resveratrol, Rapamycin and Metformin as Modulators of Antiviral Pathways. Viruses 2020, 12, 1458. https://0-doi-org.brum.beds.ac.uk/10.3390/v12121458

AMA Style

Benedetti F, Sorrenti V, Buriani A, Fortinguerra S, Scapagnini G, Zella D. Resveratrol, Rapamycin and Metformin as Modulators of Antiviral Pathways. Viruses. 2020; 12(12):1458. https://0-doi-org.brum.beds.ac.uk/10.3390/v12121458

Chicago/Turabian Style

Benedetti, Francesca, Vincenzo Sorrenti, Alessandro Buriani, Stefano Fortinguerra, Giovanni Scapagnini, and Davide Zella. 2020. "Resveratrol, Rapamycin and Metformin as Modulators of Antiviral Pathways" Viruses 12, no. 12: 1458. https://0-doi-org.brum.beds.ac.uk/10.3390/v12121458

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