Next Article in Journal
Lipid Metabolism in Macrophages: Focus on Atherosclerosis
Next Article in Special Issue
Tackling Antibiotic Resistance with Compounds of Natural Origin: A Comprehensive Review
Previous Article in Journal
Combined Treatment with Fasudil and Menthol Improves Functional Recovery in Rat Spinal Cord Injury Model
Previous Article in Special Issue
Hemocyanins from Helix and Rapana Snails Exhibit in Vitro Antitumor Effects in Human Colorectal Adenocarcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Resveratrol Modulates Transforming Growth Factor-Beta (TGF-β) Signaling Pathway for Disease Therapy: A New Insight into Its Pharmacological Activities

1
Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran
2
Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
3
Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
4
Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
5
Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417414418, Iran
6
Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956 Istanbul, Turkey
7
Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, 34956 Istanbul, Turkey
8
Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
*
Authors to whom correspondence should be addressed.
Submission received: 23 June 2020 / Revised: 28 July 2020 / Accepted: 28 July 2020 / Published: 31 July 2020
(This article belongs to the Special Issue Natural Medicine in Therapy)

Abstract

:
Resveratrol (Res) is a well-known natural product that can exhibit important pharmacological activities such as antioxidant, anti-diabetes, anti-tumor, and anti-inflammatory. An evaluation of its therapeutic effects demonstrates that this naturally occurring bioactive compound can target different molecular pathways to exert its pharmacological actions. Transforming growth factor-beta (TGF-β) is an important molecular pathway that is capable of regulating different cellular mechanisms such as proliferation, migration, and angiogenesis. TGF-β has been reported to be involved in the development of disorders such as diabetes, cancer, inflammatory disorders, fibrosis, cardiovascular disorders, etc. In the present review, the relationship between Res and TGF-β has been investigated. It was noticed that Res can inhibit TGF-β to suppress the proliferation and migration of cancer cells. In addition, Res can improve fibrosis by reducing inflammation via promoting TGF-β down-regulation. Res has been reported to be also beneficial in the amelioration of diabetic complications via targeting the TGF-β signaling pathway. These topics are discussed in detail in this review to shed light on the protective effects of Res mediated via the modulation of TGF-β signaling.

Graphical Abstract

1. Resveratrol

From immemorial times, plant-derived natural compounds have been under attention in the treatment of different disorders such as inflammatory diseases, cancers, pulmonary diseases, metabolic disorders, neurological disorders (NDs) including Alzheimer’s disease (AD) and Parkinson’s disease (PD), infertility, and so on [1,2,3,4,5,6,7,8,9,10]. Phytochemicals can exhibit beneficial actions against diseases due to their excellent pharmacological activities [11,12,13,14]. These benefits have resulted in extensive research into finding new natural compounds and revealing their potential mechanisms of actions [15,16,17]. Resveratrol (Res) is a dietary phytochemical that has been reported to be efficacious treatment for various ailments by targeting diverse molecular pathways [18,19,20,21]. The role of Res in the treatment of chronic diseases was established in early 1990s when it was found that this phytochemical possesses significant cardioprotective benefits [22]. This ascending trend toward Res research led to the revelation of its significant biological and therapeutic activities. The first report about anti-tumor activity of Res dates back to 1997, when Jang and his colleagues reported its inhibitory effect on leukemia [23].
Currently, Res can be derived from various plants including Arachis hypogea, Cassia sp., Eucalyptus sp., Morus rubra, and so on using a number of different isolation techniques [24]. High-performance liquid chromatography is the best strategy [25,26,27,28]. Over the past decades, Res has been applied in the treatment of various diseases such as osteoarthritis [29,30,31], NDs [32], cancer [33,34,35], diabetes [36], cardiovascular diseases [37], liver disorders [38], and so on. An increasing amount of evidence is in agreement with the fact that Res affects different molecular pathways to exhibit its protective effects [39,40,41]. Hence, the identification of these targets can promote further studies for investigating molecular pathways and the mechanisms of its therapeutic actions in depth. For instance, anti-inflammation is one of the most important biological effects of Res treatment. To function as an anti-inflammatory molecule, Res can effectively inhibit the activation of pro-inflammatory transcription factors such as nuclear factor-kappaB (NF-ĸB). It seems that the anti-inflammatory actions of Res are not only mediated via inhibitory actions on the NF-ĸB signaling pathway, but they also rely on its action as a PARP-γ agonist [42]. The anti-inflammatory activities of Res are also characterized by decreased levels of interleukin (IL)-6, IL-8, and tumor necrosis factor-α (TNF-α), etc. [43]. The production of pro-inflammatory lipid mediators from arachidonic acid can be mediated by the cyclooxygenase (COX) pathway. A number of anti-inflammatory drugs have been developed based on their inhibitory effect on COX-1 and COX-2 [44,45]. Res is capable of binding to the active site of COX-1 and thus causing anti-inflammatory effects. In addition to targeting inflammation, Res attaches to the active site of COX-2 to suppress cancer proliferation [46,47,48,49]. It is noteworthy that the inhibitory effect of Res on COX has been noted to follow a dose-dependent kinetics [50].
Obesity is one of the challenges faced in today’s world. Res has demonstrated great potential in reducing weight and exerting anti-obesity activity. Res changes white adipose tissue (WAT) into brown adipose tissue (BAT), which in turn decreases weight and improves insulin resistance [51]. The inhibitory action of Res on lipid accumulation leads to its effect on cardiovascular disorders. Res stimulates PARP-α/γ to activate ATP binding cassette (ABC) transporter A1/G1-mediated cholesterol efflux, resulting in a decrease in lipid accumulation and cholesterol levels. These effects can lead to a significant amelioration of atherosclerosis [52]. Based on the effect of Res on amyloid-beta (Aβ), this plant-derived natural compound is of importance in treating NDs. For instance, Res is able to inhibit inflammation and the microglial activation caused by Aβ. This results in the alleviation of inflammation (down-regulation of TNF-α and IL-6) and a diminution in apoptosis (caspase-1 down-regulation) [53]. The antioxidant activity of Res provides its protective effect during kidney injury. In rats exposed to nicotine, an increase occurs in oxidative stress markers via the down-regulation of glutathione. The administration of Res has been also correlated with improving the antioxidant defense system that protects renal cells against oxidative injury [54]. A newly published study also demonstrates the effect of Res on stem cells. Res can stimulate stem cell function to ameliorate pancreatic injury such as fibrosis and apoptosis [55]. Overall, these reports exhibit that Res has diverse therapeutic effects that have resulted in its extensive application in the treatment of various disorders [56,57,58]. In the current review, we specifically focus on the therapeutic effects of Res mediated by its regulatory action on the transforming growth factor-β (TGF-β) signaling pathway.

1.1. Resveratrol: Limitations and Applied Strategies

In spite of the excellent pharmacological activities of plant-derived natural compounds, very soon it was found that a number of issues limit their efficacy in disease treatment. Increasing evidence shows that phytochemicals are able to exert their therapeutic effects predominantly under in vitro settings. However, when their efficiency is examined for in vivo experiments, a decrease occurs in their therapeutic efficacy due to their potential poor bioavailability. The difficulty is more prominent in clinical trials, leading to a limited application of phytochemicals in clinic. This holds also true for Res, and various formulations of this agent have been tested to enhance its therapeutic capabilities. Res has a lipophilic nature and can be dissolved in fruit or vegetable juices or given in capsule form. The administration frequency of Res is variable from one to three times a day, and its reported doses are at the range of 0.073 mg to 5 g [59,60]. The reports also demonstrated that the most efficient strategy in promoting the bioavailability and protective effects of Res is using nanoparticles [61]. The encapsulation of Res by nanoparticles protects against degradation and improves its intestinal absorption and blood circulation time [62,63,64,65,66]. These benefits lead to the promoted bioavailability of Res and an improvement in its therapeutic effects [67,68]. It has been reported that loading Res on lipid carriers can significantly increase its anti-tumor activity and cytotoxicity against breast cancer cells by providing targeted delivery and enhancing its intracellular internalization [69]. Lipid nanocarriers containing Res can be administered through the oral route. The oral administration of Res-loaded lipid nanostructures is more beneficial in reducing the levels of pro-inflammatory cytokines and induction of anti-inflammatory activity compared to Res alone [70]. The enhanced release of Res in the intestine by nanoparticles is of importance in elevating its cytotoxicity against cancer cells [71]. Overall, various studies reveal that nanostructures can be considered as potential delivery systems for Res and fortunately, a significant number of studies have been performed in this field. The findings are in line with the fact that these nano-based strategies can remarkably enhance both the bioavailability and therapeutic capability of Res [72,73,74]. However, more studies are needed to design different effective nanocarriers to facilitate an optimum delivery of Res.

1.2. Pharmacokinetics of Resveratrol: A Brief Explanation

Increasing evidence demonstrates that the dosage forms and conditions of patients can affect the absorption of Res. However, the gastrointestinal (GI) tract is involved in the absorption of Res after oral administration with a peak at plasma concentration after 30 min and 1.5–2 h [75,76,77]. The absorption of Res undergoes an increase via grape consumption and using other forms such as micronized form [78,79,80,81,82]. After absorption, Res can be distributed in different organs, such as the brain, liver, intestine, and fat [83]. For metabolism, enterocytes and hepatocytes play the most important role after oral administration. Notably, Res influx occurs through the passive diffusion and carrier-mediated process [84,85]. The metabolism of Res also confirms its distribution in liver, so that it has been reported that Res is a substrate of hepatic sulfotransferase and glucuronosyltransferase, and it extensively accumulates in liver [86]. The interesting point is that metabolism of Res relies on dose. Low doses (5–50 mg) of Res are bio-transformed into glucuronides, while high doses (more than 250 mg) are bio-transformed into monosulfates [87,88,89,90,91]. Facial areas and urine are responsible for the elimination of Res. It has been noted that the administration form of Res may affect its elimination, which can be delayed when micronized Res is used [92,93,94,95,96].

1.3. Toxicity of Resveratrol

Similar to other compounds, plant-derived natural compounds have a number of drawbacks. Although Res is safe and well-tolerated at normal doses, there are toxicities associated with the application of high doses of Res [97]. The willingness toward using high doses of Res is due to its poor bioavailability, which restricts its therapeutic usage. Therefore, providing information about the toxicity of Res is advantageous for directing further studies toward using normal and safe doses of Res. It is worth mentioning that the toxicity of Res has been evaluated in both in vivo and clinical trials. It appears that high doses of Res—as much as 3 g/kg/day in rats—may result in nephrotoxicity. Although there are few studies that have demonstrated that Res can negatively affect liver and enhance levels of liver enzymes such as aspartate aminotransferase, others have reported that it may not exhibit any significant toxicity on the liver [98,99]. The administration of 750 mg/kg/day of Res for 3 months is well-tolerated in rats [100]. Studies in humans show that Res is completely safe and only a few adverse effects including blood electrolyte changes, nasopharyngitis, and erythematous rash can be observed after the administration of 400 mg of Res. Headache, myalgia, epididymitis, and dizziness were other commonly reported adverse effects of Res [101,102,103].

2. TGF-β: Signaling Pathways and Pathological Role

2.1. Members and Receptors of TGF-β Family

There are three distinct members of TGF-β in mammals including TGF-β1, TGF-β2, and TGF-β3 that are homologous in terms of structure, but they demonstrate different biological activities, temporal, and spatial expression patterns [104,105,106,107,108]. The number of genes that can encode members of the TGF-β family are numerous, but a number of them can be mentioned as activin, nodal, bone morphogenetic proteins (BMPs), and growth and differentiation factors (GDFs) [109]. The TGF-β signaling pathway possesses a regulatory effect on different cellular events such as growth, survival, differentiation, cell fate specification, angiogenesis, and so on [110,111,112,113,114,115]. TGF-β signaling is initiated by the attachment of a ligand onto cell surface receptors, which in turn triggers a cascade that mediates the translocation of TGF-β into the nucleus. In humans, there are 12 cell surface receptors that are affected by ligand, including type I receptors (ALK1-7) and type II receptors (TβRII, ActRII, ActRIIB, BMPRII, and AMHRII) [116,117]. After attachment of a certain type of TGF-β into type II receptors, these receptors are stimulated, which subsequently phosphorylates the glycine-serine-rich domain (GS domain) of type I receptors. In the canonical pathway of TGF-β, type I receptors mediate the formation of Smad complex via phosphorylation at carboxyl termini.

2.2. TGF-β Signaling Pathway

The TGF-β gene encodes a pro-precursor peptide consisting of 390 amino acids that undergoes proteolytic processing to produce mature TGF-β. This mature TGF-β has two distinct sections including amino-terminal and carboxy-terminal sections [118]. The amino-terminal fragment is known as latency associated peptide (LAP) with non-covalent attachment into TGF-β [119,120]. The cleavage of LAP by proteases or mechanical forces by cell surface integrins contributes to the release of mature and active TGF-β [121,122]. The activated TGF-β is a dimeric protein with disulfide bonds and molecular weight of 25 kDa that can bind into cell surface receptors. As described above, then, the binding of a ligand into a receptor leads to the phosphorylation of type I receptors by type II ones [123]. Then, TGF-βRI as a type I receptor can stimulate Smad2 and Smad3 via phosphorylation, resulting in the formation of a complex with Smad4. This complex translocates into the nucleus to affect target genes such as plasminogen activator inhibitor 1 (PAI1). Among them, only Smad4 and Smad3 can bind to DNA. It is worth mentioning that the affinity of Smad3 and Smad4 for attachment to DNA is low and they need to collaborate with other DNA-binding transcription factors to promote gene expression [124,125]. This is the canonical pathway of TGF-β, and there is another pathway, which is known as the non-canonical pathway. In this pathway, activated receptors target different molecular pathways such as PI3K as well as JNK, P38, extracellular signal-regulated kinase (ERK), and mitogen-activated protein kinase (MAPK). For instance, PI3K can be activated by stimulated receptors to induce Akt/mTOR axis, resulting in the stimulation of S6K and regulate protein translation (Figure 1) [106].

2.3. TGF-β in Cancer, Diabetes, and Other Pathological Events

A number of studies have highlighted that the abnormal expression of TGF-β may pave the road for generating pathological events. The role of the TGF-β signaling pathway in cancer cells has been extensively investigated. Increasing evidence demonstrates that TGF-β mediates the migration and invasion of cancer cells. For enhancing cancer cell metastasis, TGF-β induces epithelial-to-mesenchymal transition (EMT), which significantly promotes the migratory ability of cancer cells [126]. Interestingly, molecular pathways that negatively regulate the metastasis of cancer cells can reduce the expression of TGF-β. It has been revealed that sirtuin 7 (SIRT7) can suppress the migration of cancer cells through inhibiting TGF-β signaling via Smad4 degradation. Therefore, the Smad complex may be disrupted, and its nuclear translocation can be inhibited [127]. In addition to metastasis, TGF-β signaling induces angiogenesis, which is a mechanism that is vital for the proliferation and migration of cancer cells. The stimulatory effect of TGF-β on angiogenesis can be mediated via the phosphorylation of Smad3 [128]. TGF-β is able to stabilize the Nrf2 signaling pathway via p21 induction, thus leading to the chemoresistance of cancer cells [129]. Moreover, numerous studies are in agreement with the fact that TGF-β can act as a positive factor for the proliferation and migration of cancer cells, and a negative factor for cancer prognosis. In addition to cancer, TGF-β contributes to the development of other malignancies. Diabetes mellitus (DM) is a chronic metabolic disorder in which insulin resistance can be obtained and glucose metabolism undergoes dysregulation [130,131]. Myocardial injury and fibrosis may result from DM, and studies have demonstrated that TGF-β is involved in this process. In DM, TGF-β activates Smad2 to facilitate its nuclear translocation. Then, an increase occurs in fibrosis, thereby providing conditions for deteriorating DM. Mesenchymal stem cell-derived exosomes are able to improve DM fibrosis via the inhibition of the TGF-β/Smad2 axis [132]. The TGF-β/Smad3 axis may be also involved in DM fibrosis. Thus, the stimulation of TGF-β and the nuclear translocation of Smad3 provide conditions for the development of renal fibrosis during DM. It has been found that the administration of retinoic acid can alleviate DM-promoted fibrosis via the inhibition of TGF-β/Smad3 [133]. It is noteworthy that a number of phytochemicals have shown potential in the regulation of the TGF-β signaling pathway, which is of immense importance for disease therapy [134,135]. In the present review, we focus on modulation of the TGF-β signaling pathway by Res and its potential impact for disease therapy [136,137,138].

3. Resveratrol and TGF-β Signaling Pathway

In this section, we will highlight the modulatory effects of Res on TGF-β levels in different chronic diseases. For example, Res can suppress the TGF-β signaling pathway and its downstream targets such as Smads. It can also reduce TGF-β-mediated EMT in fibrosis. It has been reported that for the inhibition of EMT, Res can down-regulate matrix metalloproteinase-9 (MMP-9), leading to the alleviation of fibrosis. MicroRNAs (miRs) such as miR-31 can also be affected by Res in targeting TGF-β in disease therapy. The inhibitory effect of Res on the TGF-β signaling pathway can lead to the suppression of intra-abdominal adhesion formation, since TGF-β can enhance fibrin accumulation [139,140,141,142,143,144,145,146,147,148,149]. These modulatory effects of Res are discussed in the following sections.

3.1. Resveratrol and Fibrosis

Pulmonary fibrosis (PF) is a common disorder of the lung that is characterized with hypoxemia, restrictive functional ventilatory disturbance, and chronic fibrosis. Clinical manifestations of PF include wheezing, difficulties in breathing, and dry coughs [150]. The pathogenesis of PF is still not completely understood, but it appears that the TGF-β signaling pathway plays a significant role in PF development [151]. Thus, the administration of Res may be an ideal strategy in the amelioration of PF, and different molecular pathways may be involved. Normally, microRNA (miR)-21 can induce PF via the activation of TGF-β signaling and providing Smad7 nuclear translocation. TGF-β provides a positive feedback loop, so TGF-β enhances the expression of miR-21 and AP-1. The administration of Res down-regulates the expression of miR-21 via inhibition of the MAPK/AP-1 axis. This leads to a diminution in TGF-β expression and inhibition of Smad7, resulting in the alleviation of PF [152]. Accumulating data demonstrate that during the inhibition of fibrosis, Res affects the TGF-β signaling pathway via the modulation of miRs. Myocardial fibrosis (MF) is caused by the accumulation of collagen fibers, enhanced collagen content, and alteration in collagen composition. Systolic and diastolic functions of the heart can be negatively affected by MF [153]. TGF-β is one of the key players regulating MF [154]. The TGF-β/Smad7 axis can also contribute to the development of MF. The administration of Res can up-regulate the expression of miR-17, which in turn remarkably reduces levels of Smad7, leading to an improvement in MF [155].
In addition to PF and MF, renal fibrosis (RF) can arise as a result of the activation of the TGF-β signaling pathway. It has been reported that the inhibition of the TGF-β signaling pathway by natural products such as bardoxolone and nimbolide is of importance in RF therapy [156,157]. It is worth mentioning that Res can target the TGF-β signaling pathway, thereby causing an amelioration of RF. In RF treatment, fibroblast–myofibroblast differentiation (FMD), EMT, and the proliferation of tubular epithelial cells (TECs) should be targeted. The administration of Res can disrupt Smad2/3 activation by TGF-β and consequently suppress the proliferation of TECs, FMD, and EMT [158]. Increasing evidence demonstrates that EMT may be involved in renal fibrogenesis, and its activation can facilitate the development of RF [159,160,161,162]. Res is capable of suppressing EMT-mediated RF. It seems that TGF-β1 functions as an upstream mediator of EMT, and Res suppresses EMT and RF through inhibiting TGF-β1 [163]. In fact, in the stimulation of anti-fibrotic activity, Res affects the proliferation and survival of fibroblasts. It has been shown that Res can stimulate apoptosis in fibroblasts and suppress their growth as well. An investigation of the molecular pathways demonstrates that in targeting fibroblasts, Res can suppress TGF-β and the Smad2/3/4 complex, and it can also upregulate Smad7 [164].
It is worth mentioning that the anti-fibrotic activity of Res is dose-dependent, and using low doses is preferred as compared to higher doses. An experiment has evaluated the role of dose in the anti-fibrotic activity of Res. TGF-β induces fibrosis via formation of the Smad3/4 complex and subsequent stimulation of EMT. The administration of Res has been correlated with the deacetylation of Smad3 and Smad4 via sirtuin 1 (SIRT1). According to in vitro results, low doses of Res (5–20 mM) effectively exerted anti-fibrotic activity, while high doses (more than 40 mM) did not demonstrate any substantial anti-fibrotic activity. The in vivo findings are in line with in vitro results, so that low doses of Res (less than 25 mg/kg) improve fibrosis, while high doses of Res (more than 50 mg/kg) deteriorated the condition [165]. This study confirms the dose-related toxicity of Res. Overall, these studies demonstrate that TGF-β can function as a key player in the development of fibrosis and Res can suppress the TGF-β signaling pathway and its downstream targets such as Smads to alleviate fibrosis [166,167].
The TGF-β signaling pathway contributes to the development of fibrosis in different vital organs of body such as the lung and heart. The interesting point to highlight is the possible epigenetic regulation of TGF-β by miRs in the development of fibrosis. Res is capable of suppressing miR and TGF-β interaction in fibrosis therapy. MiR-17 and miR-21 are two important miRs that contribute to the emergence of myocardial and pulmonary fibrosis via TGF-β induction. The regulation of TGF-β by miRs is suppressed upon Res administration. RF also occurs by the function of TGF-β and subsequent induction of EMT. The TGF-β/EMT axis is inhibited by Res to alleviate RF. It is noteworthy that in the amelioration of fibrosis, components of TGF-β signaling such as Smad7 and Smad4 can also be down-regulated. Therefore, TGF-β is a versatile agent in the amelioration of fibrosis.

3.2. Resveratrol and Cancer Therapy

Accumulating data exhibit that the TGF-β signaling pathway can regulate both the proliferation and metastasis of cancer cells, and its inhibition is a promising strategy in cancer therapy [168,169,170,171,172,173]. Metastasis is an increasing challenge in the effective treatment of cancer. Cancer cells are able to migrate into neighboring and distant tissues, demanding novel strategies in the inhibition of their metastasis. EMT is one of the mechanisms that can promote invasion via the transformation of static epithelial cells into migratory mesenchymal ones [174]. A number of different molecular pathways have been recognized as regulators of EMT [175,176], and it has been found that TGF-β is capable of elevating migration via EMT induction. In breast cancer, TGF-β can stimulate EMT via Smad2 and Smad3 activation, leading to an increase in N-cadherin and vimentin levels, and a decrease in E-cadherin levels. The administration of Res suppresses the metastasis of breast cancer (under both in vitro and in vivo conditions) via the inhibition of TGF-β1 and down-regulation of Smad2 and Smad3 [177]. TGF-β also contributes to the migration and malignant behavior of lung cancer. In addition to breast cancer, Res targets TGF-β to inhibit EMT in lung cancer. By suppressing levels of TGF-β, Res down-regulates the levels of vimentin and fibronectin, while it enhances E-cadherin levels, leading to an inhibition of EMT and metastasis of lung cancer cells [178]. It is noteworthy that EMT induction enhances viability via the stimulation of cancer stem cell markers such as Bmi1 and Sox2. By inhibition of the TGF-β/Smad axis, Res not only inhibits EMT and migration, but also interferes with the proliferation and survival of cancer cells [179]. So, Res can function as a potential modulator of EMT in cancer cells to negatively affect their proliferation and metastasis.
Accumulating data also show that Res is able to diminish levels of TGF-β that in turn, suppresses the development of renal carcinoma [180]. These studies are in agreement with the fact that the inhibition of TGF-β by Res is of interest in suppressing tumor growth and metastasis [181]. Moreover, a dual relationship has been found between TGF-β and programmed cell death-1 (PD-1). For instance, PD-1 overexpression is associated with the induction of TGF-β, and TGF-β can regulate PD-1 expression [182,183]. This dual relationship is of importance in cancer therapy. Res can suppress the proliferation of oral cancer cells via the down-regulation of TGF-β and subsequent inhibition of PD-1. L-thyroxine as a thyroid hormone can also modulate the anti-tumor activity of Res via regulating the TGF-β/PD-1 axis [179].
Overall, the regulation of TGF-β by Res in cancer is of importance in terms of suppressing both migration and proliferation. The most well-known mechanism targeted by TGF-β is EMT, which can promote cancer metastasis. In addition, TGF-β can activate the signaling pathways such as PD-1 and Sox2 to ensure the growth and survival of cancer cells. Upon Res administration, TGF-β and its downstream targets are inhibited to pave the road for effective cancer therapy.

3.3. Resveratrol and Lung Injury

Injuries to vascular endothelium and alveolar epithelium by inflammatory factors can lead to the emergence of acute lung injury (ALI) [184]. Infections are able to generate ALI and among them, Pseudomonas aerogenosa, Candidate albicans, and staphylococcal enterotoxin B (SEB) are of importance [185,186,187]. In the amelioration of SEB-mediated lung injury, Res can target the TGF-β signaling pathway. Res can down-regulate the expression of miR-193a to inhibit TGF-β2 and TGFβR3, thus resulting in a decrease in levels of inflammatory cytokines and T cell infiltration [188]. The enhanced level of TGF-β has been associated with the development of asthma and lung injury [189]. In fact, the administration of Res may alleviate lung injury and asthma via decreasing levels of TGF-β [190]. Chronic obstructive pulmonary disease (COPD) is one of the most common disorders of lung tissue. Cigarette smoking is the most well-known reason for COPD [191]. Pulmonary inflammation, airflow obstruction, and remodeling are features of COPD [192]. Chronic inflammation can result in the development of COPD, and TGF-β has been found to play an important role in the pathogenesis of this disease [193,194]. Therefore, based on the modulatory impact of Res on TGF-β, the administration of this naturally occurring compound can be advantageous in the amelioration of COPD. It was also found that Res can decrease fibrotic response and inhibit mucus hypersecretion via the down-regulation of TGF-β [195].
It seems that via the regulation of TGF-β, Res is capable of reducing inflammation in lung and preventing the development of pathological events such as ALI, COPD, and asthma. Interestingly, Res inhibits inflammation via reducing the infiltration of cytokines and T cells. COPD is also emerged via pulmonary inflammation and fibrosis. Based on the effect of Res on TGF-β and subsequent decrease in fibrotic response and mucus hypersecretion, it can be beneficial in the treatment of COPD.

3.4. Resveratrol and Brain Injury

Cerebral hemorrhage is a leading cause of brain injury and vasospasm [196]. This malignancy results in ischemic/reperfusion and the induction of apoptosis in cancer cells [197,198]. The TGF-β signaling pathway has been correlated with brain injury [199]. Interestingly, the administration of Res was found to improve the blood–brain barrier (BBB) and inhibit apoptosis in neuronal cells. These protective effects of Res were found to be mediated via the inhibition of TGF-β-mediated ERK [200]. Moreover, it was found that exposing rats to alcohol is associated with an increase in levels of cytokines such as TGF-β. An administration of Res (10 and 20 mg/kg) can significantly improve cognitive deficits and reduces brain injury via decreasing TGF-β levels [201]. So, the alleviation of cognitive deficits and maintaining the integrity of BBB are functions of Res that can be mediated by TGF-β modulation.

3.5. Resveratrol and DM

During DM, microvascular complications can lead to hyperglycemia that accounts for the emergence of diabetic nephropathy (DN). Interestingly, an enhanced level of oxidative stress, renal polyol formation, protein kinase C induction, and activation of AMPK as well as the accumulation of advanced glycation end-products (AGEs) are responsible for DN [202,203]. TGF-β1 is considered as one of the potential pathways involved in the emergence of DN [204]. A combination of Res and rosuvastatin (RSU) was found to be beneficial in the alleviation of DN via the down-regulation of TGF-β1 [205]. The in vivo studies have also indicated that the administration of Res is a promising strategy in alleviating DN. It was observed that Res could diminish urinary albumin excretion, glomerular hypertrophy, and the deposition of fibronectin and collagen type IV to ameliorate DN. Moreover, an investigation of molecular pathways demonstrated that Res can alleviate TGF-β expression as well as the phosphorylation of Smad2 and Smad3 for DN alleviation (Table 1, Figure 2) [206]. The most important effect of Res during DN is reducing fibrosis, which can be mediated via TGF-β inhibition.

4. Conclusions and Future Directions

Currently, extensive research is being performed for possible applications of natural products for the therapy of chronic diseases, as these agents can regulate multiple molecular targets and transcription factors [226,227,228,229,230,231,232,233]. In the present review, a comprehensive discussion of possible impact of Res on the TGF-β signaling pathway, which is one of the important cascades involved in the regulation of biological mechanisms and the generation of pathological events, is provided. TGF-β acts as an upstream inducer of EMT, and this not only enhances the metastasis of cancer cells, but also mediates fibrosis in cells. Res inhibits TGF-β/EMT in suppressing both cancer and fibrosis. Through inhibiting TGF-β, Res diminishes the accumulation of collagen and fibrin, and reduces organ adhesion. Interestingly, Res dually targets both upstream (such as miRs) and downstream (Smads, PD-1, and EMT) mediators of TGF-β signaling in disease therapy. In addition to anti-tumor and anti-fibrotic activities, Res can also exert neuroprotective, lung protective, and anti-diabetic effects via the down-regulation of TGF-β, which was also highlighted in this article. Moreover, to circumvent the issue of poor bioavailability, the application of nanoparticles can enhance the modulatory effects of Res on the TGF-β signaling pathway. Besides, genetic manipulations such as small interfering RNA (siRNA) can also be co-applied for Res to promote its potential modulatory actions on TGF-β for therapeutic uses.
More studies are needed to find the optimal dose of Res in disease therapy via targeting TGF-β. Chemical modification of the Res structure and using nanoparticles can promote its efficacy in TGF-β regulation as well as its potential against various malignancies. More importantly, these findings are more valuable when they are translated into clinic. So, clinical studies are vital to approve the results of in vitro and in vivo experiments.

Funding

This work was also supported by a National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIP) (NRF-2018R1D1A1B07042969).

Conflicts of Interest

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

Abbreviations

NDsneurological disorders
ADAlzheimer’s disease
PDParkinson’s disease
TCMTraditional Chinese Medicine
Resresveratrol
NF-ĸBnuclear factor-kappaB
ILinterleukin
TNF-αtumor necrosis factor-α
WATwhite adipose tissue
BATbrown adipose tissue
ABCATP binding cassette
amyloid-beta
TGF-βtransforming growth factor-β
GIgastrointestinal
CPCcentrifugal partition chromatography
BMPsbone morphogenetic proteins
GDFsgrowth and differentiation factors
LAPlatency associated peptide
PAI1plasminogen activator inhibitor 1
EMTepithelial-to-mesenchymal transition
SIRT7sirtuin 7
DMdiabetes mellitus
MMP-9matrix metalloproteinase-9
PFpulmonary fibrosis
miRmicroRNA
MFmyocardial fibrosis
RFrenal fibrosis
FMDfibroblast-myofibroblast differentiation
TECstubular epithelial cells
SIRT1sirtuin 1
PD-1programmed cell death-1
ALIacute lung injury
SEBstaphylococcal enterotoxin B
COPDchronic obstructive pulmonary disease
BBBblood-brain barrier
DNdiabetic nephropathy
ERKextracellular signal-regulated kinase
MAPKmitogen-activated protein kinase
AGEsadvanced glycation end-products
RSUrosuvastatin

References

  1. Mohan, C.D.; Rangappa, S.; Preetham, H.D.; Chandra Nayaka, S.; Gupta, V.K.; Basappa, S.; Sethi, G.; Rangappa, K.S. Targeting STAT3 signaling pathway in cancer by agents derived from Mother Nature. Semin. Cancer Biol. 2020. [Google Scholar] [CrossRef] [PubMed]
  2. Aggarwal, V.; Tuli, H.S.; Thakral, F.; Singhal, P.; Aggarwal, D.; Srivastava, S.; Pandey, A.; Sak, K.; Varol, M.; Khan, M.A.; et al. Molecular mechanisms of action of hesperidin in cancer: Recent trends and advancements. Exp. Biol. Med. 2020, 245, 486–497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Baek, S.H.; Ko, J.H.; Lee, H.; Jung, J.; Kong, M.; Lee, J.W.; Lee, J.; Chinnathambi, A.; Zayed, M.E.; Alharbi, S.A.; et al. Resveratrol inhibits STAT3 signaling pathway through the induction of SOCS-1: Role in apoptosis induction and radiosensitization in head and neck tumor cells. Phytomedicine Int. J. Phytother. Phytopharm. 2016, 23, 566–577. [Google Scholar] [CrossRef] [PubMed]
  4. Dai, X.; Zhang, J.; Arfuso, F.; Chinnathambi, A.; Zayed, M.E.; Alharbi, S.A.; Kumar, A.P.; Ahn, K.S.; Sethi, G. Targeting TNF-related apoptosis-inducing ligand (TRAIL) receptor by natural products as a potential therapeutic approach for cancer therapy. Exp. Biol. Med. 2015, 240, 760–773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Prasannan, R.; Kalesh, K.A.; Shanmugam, M.K.; Nachiyappan, A.; Ramachandran, L.; Nguyen, A.H.; Kumar, A.P.; Lakshmanan, M.; Ahn, K.S.; Sethi, G. Key cell signaling pathways modulated by zerumbone: Role in the prevention and treatment of cancer. Biochem. Pharmacol. 2012, 15, 1268–1276. [Google Scholar] [CrossRef] [Green Version]
  6. Tan, S.M.; Li, F.; Rajendran, P.; Kumar, A.P.; Hui, K.M.; Sethi, G. Identification of beta-escin as a novel inhibitor of signal transducer and activator of transcription 3/Janus-activated kinase 2 signaling pathway that suppresses proliferation and induces apoptosis in human hepatocellular carcinoma cells. J. Pharmacol. Exp. Ther. 2010, 334, 285–293. [Google Scholar] [CrossRef]
  7. Lee, J.H.; Chinnathambi, A.; Alharbi, S.A.; Shair, O.H.M.; Sethi, G.; Ahn, K.S. Farnesol abrogates epithelial to mesenchymal transition process through regulating Akt/mTOR pathway. Pharmacol. Res. 2019, 150, 104504. [Google Scholar] [CrossRef]
  8. Wong, A.L.A.; Hirpara, J.L.; Pervaiz, S.; Eu, J.Q.; Sethi, G.; Goh, B.C. Do STAT3 inhibitors have potential in the future for cancer therapy? Expert Opin. Investig. Drugs 2017, 26, 883–887. [Google Scholar] [CrossRef] [Green Version]
  9. Ochiai, A.; Kuroda, K. Preconception resveratrol intake against infertility: Friend or foe? Reprod. Med. Biol. 2019, 19, 107–113. [Google Scholar] [CrossRef]
  10. McSweeney, K.R.; Gadanec, L.K.; Qaradakhi, T.; Gammune, T.M.; Kubatka, P.; Caprnda, M.; Fedotova, J.; Radonak, J.; Kruzliak, P.; Zulli, A. Impridone enhances vascular relaxation via FOXO1 pathway. Clin. Exp. Pharmacol. Physiol. 2020. [Google Scholar] [CrossRef]
  11. Kashyap, D.; Tuli, H.S.; Yerer, M.B.; Sharma, A.; Sak, K.; Srivastava, S.; Pandey, A.; Garg, V.K.; Sethi, G.; Bishayee, A. Natural product-based nanoformulations for cancer therapy: Opportunities and challenges. Semin. Cancer Biol. 2019. [Google Scholar] [CrossRef] [PubMed]
  12. Shanmugam, M.K.; Manu, K.A.; Ong, T.H.; Ramachandran, L.; Surana, R.; Bist, P.; Lim, L.H.; Kumar, A.P.; Hui, K.M.; Sethi, G. Inhibition of CXCR4/CXCL12 signaling axis by ursolic acid leads to suppression of metastasis in transgenic adenocarcinoma of mouse prostate model. Int. J. Cancer 2011, 129, 1552–1563. [Google Scholar] [CrossRef] [PubMed]
  13. Ramachandran, L.; Manu, K.A.; Shanmugam, M.K.; Li, F.; Siveen, K.S.; Vali, S.; Kapoor, S.; Abbasi, T.; Surana, R.; Smoot, D.T.; et al. Isorhamnetin inhibits proliferation and invasion and induces apoptosis through the modulation of peroxisome proliferator-activated receptor γ activation pathway in gastric cancer. J. Biol. Chem. 2012, 287, 38028–38040. [Google Scholar] [CrossRef] [Green Version]
  14. Varughese, R.S.; Lam, W.S.-T.; Marican, A.A.b.H.; Viganeshwari, S.H.; Bhave, A.S.; Syn, N.L.; Wang, J.; Wong, A.L.-A.; Kumar, A.P.; Lobie, P.E.; et al. Biopharmacological considerations for accelerating drug development of deguelin, a rotenoid with potent chemotherapeutic and chemopreventive potential. Cancer 2019, 125, 1789–1798. [Google Scholar] [CrossRef]
  15. Siveen, K.S.; Mustafa, N.; Li, F.; Kannaiyan, R.; Ahn, K.S.; Kumar, A.P.; Chng, W.J.; Sethi, G. Thymoquinone overcomes chemoresistance and enhances the anticancer effects of bortezomib through abrogation of NF-kappaB regulated gene products in multiple myeloma xenograft mouse model. Oncotarget 2014, 5, 634–648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Li, F.; Shanmugam, M.K.; Chen, L.; Chatterjee, S.; Basha, J.; Kumar, A.P.; Kundu, T.K.; Sethi, G. Garcinol, a polyisoprenylated benzophenone modulates multiple proinflammatory signaling cascades leading to the suppression of growth and survival of head and neck carcinoma. Cancer Prev. Res. 2013, 6, 843–854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Rajendran, P.; Li, F.; Shanmugam, M.K.; Vali, S.; Abbasi, T.; Kapoor, S.; Ahn, K.S.; Kumar, A.P.; Sethi, G. Honokiol inhibits signal transducer and activator of transcription-3 signaling, proliferation, and survival of hepatocellular carcinoma cells via the protein tyrosine phosphatase SHP-1. J. Cell. Physiol. 2012, 227, 2184–2195. [Google Scholar] [CrossRef] [PubMed]
  18. Huang, X.-T.; Li, X.; Xie, M.-L.; Huang, Z.; Huang, Y.-X.; Wu, G.-X.; Peng, Z.-R.; Sun, Y.-N.; Ming, Q.-L.; Liu, Y.-X. Resveratrol: Review on its discovery, anti-leukemia effects and pharmacokinetics. Chem. Biol. Interact. 2019, 306, 29–38. [Google Scholar] [CrossRef]
  19. 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] [Green Version]
  20. Shanmugam, M.K.; Warrier, S.; Kumar, A.P.; Sethi, G.; Arfuso, F. Potential Role of Natural Compounds as Anti-Angiogenic Agents in Cancer. Curr. Vasc. Pharmacol. 2017, 15, 503–519. [Google Scholar] [CrossRef]
  21. Frazzi, R.; Guardi, M. Cellular and molecular targets of resveratrol on lymphoma and leukemia cells. Molecules 2017, 22, 885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Garg, A.K.; Buchholz, T.A.; Aggarwal, B.B. Chemosensitization and radiosensitization of tumors by plant polyphenols. Antioxid. Redox Signal. 2005, 7, 1630–1647. [Google Scholar] [CrossRef]
  23. Jang, M.; Cai, L.; Udeani, G.O.; Slowing, K.V.; Thomas, C.F.; Beecher, C.W.; Fong, H.H.; Farnsworth, N.R.; Kinghorn, A.D.; Mehta, R.G. Cancer chemopreventive activity of resveratrol, a natural product derived from grapes. Science 1997, 275, 218–220. [Google Scholar] [CrossRef] [Green Version]
  24. Aggarwal, B.B.; Bhardwaj, A.; Aggarwal, R.S.; Seeram, N.P.; Shishodia, S.; Takada, Y. Role of resveratrol in prevention and therapy of cancer: Preclinical and clinical studies. Anticancer Res. 2004, 24, 2783–2840. [Google Scholar] [PubMed]
  25. Ragab, A.S.; Van Fleet, J.; Jankowski, B.; Park, J.-H.; Bobzin, S.C. Detection and quantitation of resveratrol in tomato fruit (Lycopersicon esculentum Mill.). J. Agric. Food Chem. 2006, 54, 7175–7179. [Google Scholar] [CrossRef] [PubMed]
  26. Lo, C.; Le Blanc, J.Y.; Yu, C.K.; Sze, K.; Ng, D.C.; Chu, I.K. Detection, characterization, and quantification of resveratrol glycosides in transgenic arabidopsis over-expressing a sorghum stilbene synthase gene by liquid chromatography/tandem mass spectrometry. Rapid Commun. Mass Spectrom. Int. J. Devoted Rapid Dissem. Minute Res. Mass Spectrom. 2007, 21, 4101–4108. [Google Scholar] [CrossRef]
  27. Loizzo, M.R.; Nigro, S.; De Luca, D.; Menichini, F. Detection of ochratoxin A and cis-and trans-resveratrol in red wines and their musts from Calabria (Italy). Food Addit. Contam. Part A 2011, 28, 1561–1568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Koga, C.C.; Becraft, A.R.; Lee, Y.; Lee, S.Y. Taste detection thresholds of resveratrol. J. Food Sci. 2015, 80, S2064–S2070. [Google Scholar] [CrossRef]
  29. Xu, X.; Liu, X.; Yang, Y.; He, J.; Gu, H.; Jiang, M.; Huang, Y.; Liu, X.; Liu, L. Resveratrol inhibits the development of obesity-related osteoarthritis via the TLR4 and PI3K/Akt signaling pathways. Connect. Tissue Res. 2019, 60, 571–582. [Google Scholar] [CrossRef]
  30. Ebrahim, H.A.; Alzamil, N.M.; Al-Ani, B.; Haidara, M.A.; Kamar, S.S.; Dawood, A.F. Suppression of knee joint osteoarthritis induced secondary to type 2 diabetes mellitus in rats by resveratrol: Role of glycated haemoglobin and hyperlipidaemia and biomarkers of inflammation and oxidative stress. Arch. Physiol. Biochem. 2020, 1–8. [Google Scholar] [CrossRef]
  31. Zhang, G.; Zhang, H.; You, W.; Tang, X.; Li, X.; Gong, Z. Therapeutic effect of Resveratrol in the treatment of osteoarthritis via the MALAT1/miR-9/NF-κB signaling pathway. Exp. Ther. Med. 2020, 19, 2343–2352. [Google Scholar] [CrossRef] [PubMed]
  32. Cosín-Tomàs, M.; Senserrich, J.; Arumí-Planas, M.; Alquézar, C.; Pallàs, M.; Martín-Requero, Á.; Suñol, C.; Kaliman, P.; Sanfeliu, C. Role of Resveratrol and Selenium on Oxidative Stress and Expression of Antioxidant and Anti-Aging Genes in Immortalized Lymphocytes from Alzheimer’s Disease Patients. Nutrients 2019, 11, 1764. [Google Scholar] [CrossRef] [Green Version]
  33. Yuan, L.; Zhou, M.; Huang, D.; Wasan, H.S.; Zhang, K.; Sun, L.; Huang, H.; Ma, S.; Shen, M.; Ruan, S. Resveratrol inhibits the invasion and metastasis of colon cancer through reversal of epithelial- mesenchymal transition via the AKT/GSK-3β/Snail signaling pathway. Mol. Med. Rep. 2019, 20, 2783–2795. [Google Scholar] [CrossRef] [PubMed]
  34. Jang, Y.G.; Go, R.E.; Hwang, K.A.; Choi, K.C. Resveratrol inhibits DHT-induced progression of prostate cancer cell line through interfering with the AR and CXCR4 pathway. J. Steroid Biochem. Mol. Biol. 2019, 192, 105406. [Google Scholar] [CrossRef] [PubMed]
  35. Kiskova, T.; Kubatka, P.; Büsselberg, D.; Kassayova, M. The Plant-Derived Compound Resveratrol in Brain Cancer: A Review. Biomolecules 2020, 10, 161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Rašković, A.; Ćućuz, V.; Torović, L.; Tomas, A.; Gojković-Bukarica, L.; Ćebović, T.; Milijašević, B.; Stilinović, N.; Cvejić Hogervorst, J. Resveratrol supplementation improves metabolic control in rats with induced hyperlipidemia and type 2 diabetes. Saudi Pharm. J. SPJ Off. Publ. Saudi Pharm. Soc. 2019, 27, 1036–1043. [Google Scholar] [CrossRef]
  37. Hong, M.; Li, J.; Li, S.; Almutairi, M.M. Resveratrol Derivative, Trans-3, 5, 4′-Trimethoxystilbene, Prevents the Developing of Atherosclerotic Lesions and Attenuates Cholesterol Accumulation in Macrophage Foam Cells. Mol. Nutr. Food Res. 2020, 64, e1901115. [Google Scholar] [CrossRef]
  38. Yu, B.; Qin, S.Y.; Hu, B.L.; Qin, Q.Y.; Jiang, H.X.; Luo, W. Resveratrol improves CCL4-induced liver fibrosis in mouse by upregulating endogenous IL-10 to reprogramme macrophages phenotype from M(LPS) to M(IL-4). Biomed. Pharmacother. 2019, 117, 109110. [Google Scholar] [CrossRef]
  39. Tewari, D.; Nabavi, S.F.; Nabavi, S.M.; Sureda, A.; Farooqi, A.A.; Atanasov, A.G.; Vacca, R.A.; Sethi, G.; Bishayee, A. Targeting activator protein 1 signaling pathway by bioactive natural agents: Possible therapeutic strategy for cancer prevention and intervention. Pharm. Res. 2018, 128, 366–375. [Google Scholar] [CrossRef]
  40. Deng, S.; Shanmugam, M.K.; Kumar, A.P.; Yap, C.T.; Sethi, G.; Bishayee, A. Targeting autophagy using natural compounds for cancer prevention and therapy. Cancer 2019, 125, 1228–1246. [Google Scholar] [CrossRef]
  41. Mishra, S.; Verma, S.S.; Rai, V.; Awasthee, N.; Chava, S.; Hui, K.M.; Kumar, A.P.; Challagundla, K.B.; Sethi, G.; Gupta, S.C. Long non-coding RNAs are emerging targets of phytochemicals for cancer and other chronic diseases. Cell. Mol. Life Sci. CMLS 2019, 76, 1947–1966. [Google Scholar] [CrossRef] [PubMed]
  42. Ben Lagha, A.; Andrian, E.; Grenier, D. Resveratrol attenuates the pathogenic and inflammatory properties of Porphyromonas gingivalis. Mol. Oral Microbiol. 2019, 34, 118–130. [Google Scholar] [CrossRef] [PubMed]
  43. Farzanegan, A.; Shokuhian, M.; Jafari, S.; Shirazi, F.S.; Shahidi, M. Anti-histaminic Effects of Resveratrol and Silymarin on Human Gingival Fibroblasts. Inflammation 2019, 42, 1622–1629. [Google Scholar] [CrossRef] [PubMed]
  44. Calamini, B.; Ratia, K.; Malkowski, M.G.; Cuendet, M.; Pezzuto, J.M.; Santarsiero, B.D.; Mesecar, A.D. Pleiotropic mechanisms facilitated by resveratrol and its metabolites. Biochem. J. 2010, 429, 273–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Hwang, S.H.; Wecksler, A.T.; Wagner, K.; Hammock, B.D. Rationally designed multitarget agents against inflammation and pain. Curr. Med. Chem. 2013, 20, 1783–1799. [Google Scholar] [CrossRef] [PubMed]
  46. Lançon, A.; Frazzi, R.; Latruffe, N. Anti-oxidant, anti-inflammatory and anti-angiogenic properties of resveratrol in ocular diseases. Molecules 2016, 21, 304. [Google Scholar] [CrossRef]
  47. Cheng, T.M.; Chin, Y.T.; Ho, Y.; Chen, Y.R.; Yang, Y.N.; Yang, Y.C.; Shih, Y.J.; Lin, T.I.; Lin, H.Y.; Davis, P.J. Resveratrol induces sumoylated COX-2-dependent anti-proliferation in human prostate cancer LNCaP cells. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2018, 112, 67–75. [Google Scholar] [CrossRef]
  48. Gong, W.H.; Zhao, N.; Zhang, Z.M.; Zhang, Y.X.; Yan, L.; Li, J.B. The inhibitory effect of resveratrol on COX-2 expression in human colorectal cancer: A promising therapeutic strategy. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 1136–1143. [Google Scholar]
  49. Zykova, T.A.; Zhu, F.; Zhai, X.; Ma, W.Y.; Ermakova, S.P.; Lee, K.W.; Bode, A.M.; Dong, Z. Resveratrol directly targets COX-2 to inhibit carcinogenesis. Mol. Carcinog. 2008, 47, 797–805. [Google Scholar] [CrossRef] [Green Version]
  50. Latruffe, N.; Lançon, A.; Frazzi, R.; Aires, V.; Delmas, D.; 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]
  51. Kim, O.Y.; Chung, J.Y.; Song, J. Effect of resveratrol on adipokines and myokines involved in fat browning: Perspectives in healthy weight against obesity. Pharm. Res. 2019, 148, 104411. [Google Scholar] [CrossRef] [PubMed]
  52. Ye, G.; Chen, G.; Gao, H.; Lin, Y.; Liao, X.; Zhang, H.; Liu, X.; Chi, Y.; Huang, Q.; Zhu, H.; et al. Resveratrol inhibits lipid accumulation in the intestine of atherosclerotic mice and macrophages. J. Cell. Mol. Med. 2019, 23, 4313–4325. [Google Scholar] [CrossRef] [PubMed]
  53. Feng, L.; Zhang, L. Resveratrol Suppresses Aβ-Induced Microglial Activation Through the TXNIP/TRX/NLRP3 Signaling Pathway. DNA Cell Biol. 2019, 38, 874–879. [Google Scholar] [CrossRef] [PubMed]
  54. Ramalingam, A.; Santhanathas, T.; Shaukat Ali, S.; Zainalabidin, S. Resveratrol Supplementation Protects Against Nicotine-Induced Kidney Injury. Int. J. Environ. Res. Public Health 2019, 16, 4445. [Google Scholar] [CrossRef] [Green Version]
  55. Chen, T.S.; Kuo, C.H.; Day, C.H.; Pan, L.F.; Chen, R.J.; Chen, B.C.; Padma, V.V.; Lin, Y.M.; Huang, C.Y. Resveratrol increases stem cell function in the treatment of damaged pancreas. J. Cell. Physiol. 2019, 234, 20443–20452. [Google Scholar] [CrossRef]
  56. Wang, Y.; Wang, B.; Qi, X.; Zhang, X.; Ren, K. Resveratrol Protects Against Post-Contrast Acute Kidney Injury in Rabbits With Diabetic Nephropathy. Front. Pharmacol. 2019, 10, 833. [Google Scholar] [CrossRef] [Green Version]
  57. Lieben Louis, X.; Raj, P.; Meikle, Z.; Yu, L.; Susser, S.E.; MacInnis, S.; Duhamel, T.A.; Wigle, J.T.; Netticadan, T. Resveratrol prevents palmitic-acid-induced cardiomyocyte contractile impairment. Can. J. Physiol. Pharmacol. 2019, 97, 1132–1140. [Google Scholar] [CrossRef]
  58. Gimeno-Mallench, L.; Mas-Bargues, C.; Inglés, M.; Olaso, G.; Borras, C.; Gambini, J.; Vina, J. Resveratrol shifts energy metabolism to increase lipid oxidation in healthy old mice. Biomed. Pharmacother. 2019, 118, 109130. [Google Scholar] [CrossRef]
  59. Amri, A.; Chaumeil, J.; Sfar, S.; Charrueau, C. Administration of resveratrol: What formulation solutions to bioavailability limitations? J. Control. Release 2012, 158, 182–193. [Google Scholar] [CrossRef]
  60. Chauhan, A.S. Dendrimer nanotechnology for enhanced formulation and controlled delivery of resveratrol. Ann. N. Y. Acad. Sci. 2015, 1348, 134–140. [Google Scholar] [CrossRef]
  61. Santos, A.C.; Pereira, I.; Pereira-Silva, M.; Ferreira, L.; Caldas, M.; Collado-González, M.; Magalhães, M.; Figueiras, A.; Ribeiro, A.J.; Veiga, F. Nanotechnology-based formulations for resveratrol delivery: Effects on resveratrol in vivo bioavailability and bioactivity. Colloids Surf. B Biointerfaces 2019, 180, 127–140. [Google Scholar] [CrossRef] [PubMed]
  62. Huang, M.; Liang, C.; Tan, C.; Huang, S.; Ying, R.; Wang, Y.; Wang, Z.; Zhang, Y. Liposome co-encapsulation as a strategy for the delivery of curcumin and resveratrol. Food Funct. 2019, 10, 6447–6458. [Google Scholar] [CrossRef] [PubMed]
  63. Ravikumar, P.; Katariya, M.; Patil, S.; Tatke, P.; Pillai, R. Skin delivery of resveratrol encapsulated lipidic formulation for melanoma chemoprevention. J. Microencapsul. 2019, 36, 535–551. [Google Scholar] [CrossRef] [PubMed]
  64. Intagliata, S.; Modica, M.N.; Santagati, L.M.; Montenegro, L. Strategies to Improve Resveratrol Systemic and Topical Bioavailability: An Update. Antioxidants 2019, 8, 244. [Google Scholar] [CrossRef] [Green Version]
  65. Hu, Y.; Wang, Z.; Qiu, Y.; Liu, Y.; Ding, M.; Zhang, Y. Anti-miRNA21 and resveratrol-loaded polysaccharide-based mesoporous silica nanoparticle for synergistic activity in gastric carcinoma. J. Drug Target. 2019, 27, 1135–1143. [Google Scholar] [CrossRef]
  66. Machado, N.D.; Fernández, M.A.; Díaz, D.D. Recent Strategies in Resveratrol Delivery Systems. ChemPlusChem 2019, 84, 951–973. [Google Scholar] [CrossRef]
  67. Ruginǎ, D.; Ghiman, R.; Focșan, M.; Tăbăran, F.; Copaciu, F.; Suciu, M.; Pintea, A.; Aștilean, S. Resveratrol-delivery vehicle with anti-VEGF activity carried to human retinal pigmented epithelial cells exposed to high-glucose induced conditions. Colloids Surf. B Biointerfaces 2019, 181, 66–75. [Google Scholar] [CrossRef]
  68. Kang, J.H.; Ko, Y.T. Enhanced Subcellular Trafficking of Resveratrol Using Mitochondriotropic Liposomes in Cancer Cells. Pharmaceutics 2019, 11, 423. [Google Scholar] [CrossRef] [Green Version]
  69. Poonia, N.; Kaur Narang, J.; Lather, V.; Beg, S.; Sharma, T.; Singh, B.; Pandita, D. Resveratrol loaded functionalized nanostructured lipid carriers for breast cancer targeting: Systematic development, characterization and pharmacokinetic evaluation. Colloids Surf. B Biointerfaces 2019, 181, 756–766. [Google Scholar] [CrossRef]
  70. de Oliveira, M.T.P.; de Sá Coutinho, D.; Tenório de Souza, É.; Stanisçuaski Guterres, S.; Pohlmann, A.R.; Silva, P.M.R.; Martins, M.A.; Bernardi, A. Orally delivered resveratrol-loaded lipid-core nanocapsules ameliorate LPS-induced acute lung injury via the ERK and PI3K/Akt pathways. Int. J. Nanomed. 2019, 14, 5215–5228. [Google Scholar] [CrossRef] [Green Version]
  71. Rostami, M.; Ghorbani, M.; Aman Mohammadi, M.; Delavar, M.; Tabibiazar, M.; Ramezani, S. Development of resveratrol loaded chitosan-gellan nanofiber as a novel gastrointestinal delivery system. Int. J. Biol. Macromol. 2019, 135, 698–705. [Google Scholar] [CrossRef] [PubMed]
  72. Yang, C.; Wang, Y.; Xie, Y.; Liu, G.; Lu, Y.; Wu, W.; Chen, L. Oat protein-shellac nanoparticles as a delivery vehicle for resveratrol to improve bioavailability in vitro and in vivo. Nanomedicine 2019, 14, 2853–2871. [Google Scholar] [CrossRef] [PubMed]
  73. Ha, E.S.; Sim, W.Y.; Lee, S.K.; Jeong, J.S.; Kim, J.S.; Baek, I.H.; Choi, D.H.; Park, H.; Hwang, S.J.; Kim, M.S. Preparation and Evaluation of Resveratrol-Loaded Composite Nanoparticles Using a Supercritical Fluid Technology for Enhanced Oral and Skin Delivery. Antioxidants 2019, 8, 554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Sharma, B.; Iqbal, B.; Kumar, S.; Ali, J.; Baboota, S. Resveratrol-loaded nanoemulsion gel system to ameliorate UV-induced oxidative skin damage: From in vitro to in vivo investigation of antioxidant activity enhancement. Arch. Dermatol. Res. 2019, 311, 773–793. [Google Scholar] [CrossRef] [PubMed]
  75. Chukwumah, Y.; Walker, L.; Vogler, B.; Verghese, M. In vitro absorption of dietary trans-resveratrol from boiled and roasted peanuts in Caco-2 cells. J. Agric. Food Chem. 2011, 59, 12323–12329. [Google Scholar] [CrossRef]
  76. Soleas, G.J.; Angelini, M.; Grass, L.; Diamandis, E.P.; Goldberg, D.M. Absorption of trans-resveratrol in rats. In Methods in Enzymology; Elsevier: Amsterdam, The Netherlands, 2001; Volume 335, pp. 145–154. [Google Scholar]
  77. Willenberg, I.; Michael, M.; Wonik, J.; Bartel, L.C.; Empl, M.T.; Schebb, N.H. Investigation of the absorption of resveratrol oligomers in the Caco-2 cellular model of intestinal absorption. Food Chem. 2015, 167, 245–250. [Google Scholar] [CrossRef]
  78. 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] [Green Version]
  79. Delmas, D.; Aires, V.; Colin, D.J.; Limagne, E.; Scagliarini, A.; Cotte, A.K.; Ghiringhelli, F. Importance of lipid microdomains, rafts, in absorption, delivery, and biological effects of resveratrol. Ann. N. Y. Acad. Sci. 2013, 1290, 90–97. [Google Scholar] [CrossRef]
  80. Polonini, H.C.; de Almeida Bastos, C.; de Oliveira, M.A.L.; da Silva, C.G.A.; Collins, C.H.; Brandão, M.A.F.; Raposo, N.R.B. In vitro drug release and ex vivo percutaneous absorption of resveratrol cream using HPLC with zirconized silica stationary phase. J. Chromatogr. B 2014, 947, 23–31. [Google Scholar] [CrossRef]
  81. Biasutto, L.; Marotta, E.; Mattarei, A.; Beltramello, S.; Caliceti, P.; Salmaso, S.; Bernkop-Schnürch, A.; Garbisa, S.; Zoratti, M.; Paradisi, C. Absorption and metabolism of resveratrol carboxyesters and methanesulfonate by explanted rat intestinal segments. Cell. Physiol. Biochem. 2009, 24, 557–566. [Google Scholar] [CrossRef]
  82. Basavaraj, S.; Betageri, G.V. Improved oral delivery of resveratrol using proliposomal formulation: Investigation of various factors contributing to prolonged absorption of unmetabolized resveratrol. Expert Opin. Drug Deliv. 2014, 11, 493–503. [Google Scholar] [CrossRef] [PubMed]
  83. Andres-Lacueva, C.; Macarulla, M.T.; Rotches-Ribalta, M.; Boto-Ordóñez, M.; Urpi-Sarda, M.; Rodríguez, V.M.; Portillo, M.P. Distribution of resveratrol metabolites in liver, adipose tissue, and skeletal muscle in rats fed different doses of this polyphenol. J. Agric. Food Chem. 2012, 60, 4833–4840. [Google Scholar] [CrossRef]
  84. Bertelli, A.; Baccalini, R.; Battaglia, E.; Falchi, M.; Ferrero, M. Resveratrol inhibits TNF alpha-induced endothelial cell activation. Therapie 2001, 56, 613–616. [Google Scholar] [PubMed]
  85. Lançon, A.; Hanet, N.; Jannin, B.; Delmas, D.; Heydel, J.-M.; Lizard, G.; Chagnon, M.-C.; Artur, Y.; Latruffe, N. Resveratrol in human hepatoma HepG2 cells: Metabolism and inducibility of detoxifying enzymes. Drug Metab. Dispos. 2007, 35, 699–703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. De Santi, C.; Pietrabissa, A.; Spisni, R.; Mosca, F.; Pacifici, G. Sulphation of resveratrol, a natural product present in grapes and wine, in the human liver and duodenum. Xenobiotica 2000, 30, 609–617. [Google Scholar] [CrossRef] [PubMed]
  87. Murias, M.; Miksits, M.; Aust, S.; Spatzenegger, M.; Thalhammer, T.; Szekeres, T.; Jaeger, W. Metabolism of resveratrol in breast cancer cell lines: Impact of sulfotransferase 1A1 expression on cell growth inhibition. Cancer Lett. 2008, 261, 172–182. [Google Scholar] [CrossRef]
  88. Azorín-Ortuño, M.; Yáñez-Gascón, M.J.; Vallejo, F.; Pallarés, F.J.; Larrosa, M.; Lucas, R.; Morales, J.C.; Tomás-Barberán, F.A.; García-Conesa, M.T.; Espín, J.C. Metabolites and tissue distribution of resveratrol in the pig. Mol. Nutr. Food Res. 2011, 55, 1154–1168. [Google Scholar] [CrossRef] [Green Version]
  89. Bode, L.M.; Bunzel, D.; Huch, M.; Cho, G.-S.; Ruhland, D.; Bunzel, M.; Bub, A.; Franz, C.M.; Kulling, S.E. In vivo and in vitro metabolism of trans-resveratrol by human gut microbiota. Am. J. Clin. Nutr. 2013, 97, 295–309. [Google Scholar] [CrossRef]
  90. El-Sherbeni, A.A.; El-Kadi, A.O. Characterization of arachidonic acid metabolism by rat cytochrome P450 enzymes: The involvement of CYP1As. Drug Metab. Dispos. 2014, 42, 1498–1507. [Google Scholar] [CrossRef] [Green Version]
  91. Xiao, X.; Wu, Z.-C.; Chou, K.-C. A multi-label classifier for predicting the subcellular localization of gram-negative bacterial proteins with both single and multiple sites. PLoS ONE 2011, 6, e20592. [Google Scholar] [CrossRef] [Green Version]
  92. Ortuño, J.; Covas, M.-I.; Farre, M.; Pujadas, M.; Fito, M.; Khymenets, O.; Andres-Lacueva, C.; Roset, P.; Joglar, J.; Lamuela-Raventós, R.M. Matrix effects on the bioavailability of resveratrol in humans. Food Chem. 2010, 120, 1123–1130. [Google Scholar] [CrossRef]
  93. Rotches-Ribalta, M.; Andres-Lacueva, C.; Estruch, R.; Escribano, E.; Urpi-Sarda, M. Pharmacokinetics of resveratrol metabolic profile in healthy humans after moderate consumption of red wine and grape extract tablets. Pharmacol. Res. 2012, 66, 375–382. [Google Scholar] [CrossRef] [PubMed]
  94. Vickers, N.J. Animal Communication: When I’m Calling You, Will You Answer Too? Curr. Biol. 2017, 27, R713–R715. [Google Scholar] [CrossRef] [PubMed]
  95. De Bock, M.; Thorstensen, E.B.; Derraik, J.G.; Henderson, H.V.; Hofman, P.L.; Cutfield, W.S. Human absorption and metabolism of oleuropein and hydroxytyrosol ingested as olive (O lea europaea L.) leaf extract. Mol. Nutr. Food Res. 2013, 57, 2079–2085. [Google Scholar] [CrossRef]
  96. Menet, M.-C.; Marchal, J.; Dal-Pan, A.; Taghi, M.; Nivet-Antoine, V.; Dargère, D.; Laprévote, O.; Beaudeux, J.-L.; Aujard, F.; Epelbaum, J. Resveratrol Metabolism in a Non-Human Primate, the Grey Mouse Lemur (Microcebus murinus), Using Ultra-High-Performance Liquid Chromatography–Quadrupole Time of Flight. PLoS ONE 2014, 9, e91932. [Google Scholar] [CrossRef]
  97. Cottart, C.H.; Nivet-Antoine, V.; Laguillier-Morizot, C.; Beaudeux, J.L. Resveratrol bioavailability and toxicity in humans. Mol. Nutr. Food Res. 2010, 54, 7–16. [Google Scholar] [CrossRef]
  98. Crowell, J.A.; Korytko, P.J.; Morrissey, R.L.; Booth, T.D.; Levine, B.S. Resveratrol-associated renal toxicity. Toxicol. Sci. 2004, 82, 614–619. [Google Scholar] [CrossRef] [Green Version]
  99. Juan, M.E.; Vinardell, M.P.; Planas, J.M. The daily oral administration of high doses of trans-resveratrol to rats for 28 days is not harmful. J. Nutr. 2002, 132, 257–260. [Google Scholar] [CrossRef] [Green Version]
  100. Williams, L.D.; Burdock, G.A.; Edwards, J.A.; Beck, M.; Bausch, J. Safety studies conducted on high-purity trans-resveratrol in experimental animals. Food Chem. Toxicol. 2009, 47, 2170–2182. [Google Scholar] [CrossRef]
  101. Boocock, D.J.; Faust, G.E.; Patel, K.R.; Schinas, A.M.; Brown, V.A.; Ducharme, M.P.; Booth, T.D.; Crowell, J.A.; Perloff, M.; Gescher, A.J. Phase I dose escalation pharmacokinetic study in healthy volunteers of resveratrol, a potential cancer chemopreventive agent. Cancer Epidemiol. Prev. Biomark. 2007, 16, 1246–1252. [Google Scholar] [CrossRef] [Green Version]
  102. Vaz-da-Silva, M.; Loureiro, A.; Falcao, A.; Nunes, T.; Rocha, J.; Fernandes-Lopes, C.; Soares, E.; Wright, L.; Almeida, L.; Soares-da-Silva, P. Effect of food on the pharmacokinetic profile of trans-resveratrol. Int. J. Clin. Pharm. 2008, 46, 564–570. [Google Scholar] [CrossRef]
  103. Almeida, L.; Vaz-da-Silva, M.; Falcão, A.; Soares, E.; Costa, R.; Loureiro, A.I.; Fernandes-Lopes, C.; Rocha, J.F.; Nunes, T.; Wright, L. Pharmacokinetic and safety profile of trans-resveratrol in a rising multiple-dose study in healthy volunteers. Mol. Nutr. Food Res. 2009, 53, S7–S15. [Google Scholar] [CrossRef] [PubMed]
  104. Hao, Y.; Baker, D.; ten Dijke, P. TGF-β-mediated epithelial-mesenchymal transition and cancer metastasis. Int. J. Mol. Sci. 2019, 20, 2767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Boguslawska, J.; Kryst, P.; Poletajew, S.; Piekielko-Witkowska, A. TGF-β and microRNA Interplay in Genitourinary Cancers. Cells 2019, 8, 1619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Colak, S.; ten Dijke, P. Targeting TGF-β signaling in cancer. Trends Cancer 2017, 3, 56–71. [Google Scholar] [CrossRef]
  107. Van Der Kraan, P.M. The changing role of TGFβ in healthy, ageing and osteoarthritic joints. Nat. Rev. Rheumatol. 2017, 13, 155. [Google Scholar] [CrossRef]
  108. Chen, S.; Liu, S.; Ma, K.; Zhao, L.; Lin, H.; Shao, Z. TGF-β signaling in intervertebral disc health and disease. Osteoarthr. Cartil. 2019, 27, 1109–1117. [Google Scholar] [CrossRef]
  109. Yu, Y.; Feng, X.-H. TGF-β signaling in cell fate control and cancer. Curr. Opin. Cell Biol. 2019, 61, 56–63. [Google Scholar] [CrossRef]
  110. Chung, C.-L.; Tai, S.-B.; Hu, T.-H.; Chen, J.-J.; Chen, C.-L. Roles of Myosin-Mediated Membrane Trafficking in TGF-β Signaling. Int. J. Mol. Sci. 2019, 20, 3913. [Google Scholar] [CrossRef] [Green Version]
  111. Muñoz, M.; Sánchez-Capelo, A. TGF-β/Smad3 Signalling Modulates GABA Neurotransmission: Implications in Parkinson’s Disease. Int. J. Mol. Sci. 2020, 21, 590. [Google Scholar] [CrossRef] [Green Version]
  112. Liarte, S.; Bernabé-García, Á.; Nicolás, F.J. Role of TGF-in Skin Chronic Wounds: A Keratinocyte Perspective. Cells 2020, 9, 306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Samarakoon, R.; Higgins, S.P.; Higgins, C.E.; Higgins, P.J. The TGF-β1/p53/PAI-1 Signaling Axis in Vascular Senescence: Role of Caveolin-1. Biomolecules 2019, 9, 341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Tzavlaki, K.; Moustakas, A. TGF-β Signaling. Biomolecules 2020, 10, 487. [Google Scholar] [CrossRef] [Green Version]
  115. Suriyamurthy, S.; Baker, D.; ten Dijke, P.; Iyengar, P.V. Epigenetic reprogramming of TGF-β signaling in breast cancer. Cancers 2019, 11, 726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Massagué, J. TGFβ signalling in context. Nat. Rev. Mol. Cell Biol. 2012, 13, 616–630. [Google Scholar] [CrossRef]
  117. Derynck, R.; Miyazono, K. The Biology of the TGF-β Family; Cold Spring Harbor Laboratory Press: New York, NY, USA, 2017. [Google Scholar]
  118. Derynck, R.; Jarrett, J.A.; Chen, E.Y.; Eaton, D.H.; Bell, J.R.; Assoian, R.K.; Roberts, A.B.; Sporn, M.B.; Goeddel, D.V. Human transforming growth factor-β complementary DNA sequence and expression in normal and transformed cells. Nature 1985, 316, 701–705. [Google Scholar] [CrossRef] [Green Version]
  119. Sha, X.; Yang, L.; Gentry, L.E. Identification and analysis of discrete functional domains in the pro region of pre-pro-transforming growth factor beta 1. J. Cell Biol. 1991, 114, 827–839. [Google Scholar] [CrossRef] [Green Version]
  120. Shi, M.; Zhu, J.; Wang, R.; Chen, X.; Mi, L.; Walz, T.; Springer, T.A. Latent TGF-β structure and activation. Nature 2011, 474, 343–349. [Google Scholar] [CrossRef] [Green Version]
  121. Cheifetz, S.; Hernandez, H.; Laiho, M.; Ten Dijke, P.; Iwata, K.K.; Massagué, J. Distinct transforming growth factor-beta (TGF-beta) receptor subsets as determinants of cellular responsiveness to three TGF-beta isoforms. J. Biol. Chem. 1990, 265, 20533–20538. [Google Scholar]
  122. Dong, X.; Zhao, B.; Iacob, R.E.; Zhu, J.; Koksal, A.C.; Lu, C.; Engen, J.R.; Springer, T.A. Force interacts with macromolecular structure in activation of TGF-β. Nature 2017, 542, 55–59. [Google Scholar] [CrossRef]
  123. Marafini, I.; Troncone, E.; Salvatori, S.; Monteleone, G. TGF-β activity restoration and phosphodiesterase 4 inhibition as therapeutic options for inflammatory bowel diseases. Pharmacol. Res. 2020, 104757. [Google Scholar] [CrossRef] [PubMed]
  124. Heldin, C.-H.; Moustakas, A. Role of Smads in TGFβ signaling. Cell Tissue Res. 2012, 347, 21–36. [Google Scholar] [CrossRef]
  125. Schmierer, B.; Hill, C.S. TGFβ–SMAD signal transduction: Molecular specificity and functional flexibility. Nat. Rev. Mol. Cell Biol. 2007, 8, 970–982. [Google Scholar] [CrossRef]
  126. Zhang, L.; Zhou, F.; García de Vinuesa, A.; de Kruijf, E.M.; Mesker, W.E.; Hui, L.; Drabsch, Y.; Li, Y.; Bauer, A.; Rousseau, A.; et al. TRAF4 promotes TGF-β receptor signaling and drives breast cancer metastasis. Mol. Cell 2013, 51, 559–572. [Google Scholar] [CrossRef] [Green Version]
  127. Tang, X.; Shi, L.; Xie, N.; Liu, Z.; Qian, M.; Meng, F.; Xu, Q.; Zhou, M.; Cao, X.; Zhu, W.G.; et al. SIRT7 antagonizes TGF-β signaling and inhibits breast cancer metastasis. Nat. Commun. 2017, 8, 318. [Google Scholar] [CrossRef] [Green Version]
  128. Muppala, S.; Xiao, R.; Krukovets, I.; Verbovetsky, D.; Yendamuri, R.; Habib, N.; Raman, P.; Plow, E.; Stenina-Adognravi, O. Thrombospondin-4 mediates TGF-β-induced angiogenesis. Oncogene 2017, 36, 5189–5198. [Google Scholar] [CrossRef] [Green Version]
  129. Oshimori, N.; Oristian, D.; Fuchs, E. TGF-β promotes heterogeneity and drug resistance in squamous cell carcinoma. Cell 2015, 160, 963–976. [Google Scholar] [CrossRef] [Green Version]
  130. Movahed, A.; Raj, P.; Nabipour, I.; Mahmoodi, M.; Ostovar, A.; Kalantarhormozi, M.; Netticadan, T. Efficacy and Safety of Resveratrol in Type 1 Diabetes Patients: A Two-Month Preliminary Exploratory Trial. Nutrients 2020, 12, 161. [Google Scholar] [CrossRef] [Green Version]
  131. Bahmanzadeh, M.; Goodarzi, M.T.; Rezaei Farimani, A.; Fathi, N.; Alizadeh, Z. Resveratrol supplementation improves DNA integrity and sperm parameters in streptozotocin-nicotinamide-induced type 2 diabetic rats. Andrologia 2019, 51, e13313. [Google Scholar] [CrossRef]
  132. Lin, Y.; Zhang, F.; Lian, X.F.; Peng, W.Q.; Yin, C.Y. Mesenchymal stem cell-derived exosomes improve diabetes mellitus-induced myocardial injury and fibrosis via inhibition of TGF-β1/Smad2 signaling pathway. Cell. Mol. Biol. 2019, 65, 123–126. [Google Scholar] [CrossRef]
  133. Sierra-Mondragon, E.; Rodríguez-Muñoz, R.; Namorado-Tonix, C.; Molina-Jijon, E.; Romero-Trejo, D.; Pedraza-Chaverri, J.; Reyes, J.L. All-Trans Retinoic Acid Attenuates Fibrotic Processes by Downregulating TGF-β1/Smad3 in Early Diabetic Nephropathy. Biomolecules 2019, 9, 525. [Google Scholar] [CrossRef] [Green Version]
  134. Hsu, W.H.; Liao, S.C.; Chyan, Y.J.; Huang, K.W.; Hsu, S.L.; Chen, Y.C.; Siu, M.L.; Chang, C.C.; Chung, Y.S.; Huang, C.F. Graptopetalum paraguayense Inhibits Liver Fibrosis by Blocking TGF-β Signaling In Vivo and In Vitro. Int. J. Mol. Sci. 2019, 20, 2592. [Google Scholar] [CrossRef] [Green Version]
  135. Ma, J.Q.; Sun, Y.Z.; Ming, Q.L.; Tian, Z.K.; Yang, H.X.; Liu, C.M. Ampelopsin attenuates carbon tetrachloride-induced mouse liver fibrosis and hepatic stellate cell activation associated with the SIRT1/TGF-β1/Smad3 and autophagy pathway. Int. Immunopharmacol. 2019, 77, 105984. [Google Scholar] [CrossRef]
  136. Razali, N.; Agarwal, R.; Agarwal, P.; Froemming, G.R.A.; Tripathy, M.; Ismail, N.M. IOP lowering effect of topical trans-resveratrol involves adenosine receptors and TGF-β2 signaling pathways. Eur. J. Pharmacol. 2018, 838, 1–10. [Google Scholar] [CrossRef]
  137. Yang, R.C.; Zhu, X.L.; Zhang, H.Q.; Li, W.D. Study of resveratrol suppressing TGF-beta1 induced transdifferentiation of podocytes. Zhongguo Zhong Xi Yi Jie He Za Zhi Zhongguo Zhongxiyi Jiehe Zazhi Chin. J. Integr. Tradit. West. Med. 2013, 33, 1677–1682. [Google Scholar]
  138. Suenaga, F.; Hatsushika, K.; Takano, S.; Ando, T.; Ohnuma, Y.; Ogawa, H.; Nakao, A. A possible link between resveratrol and TGF-beta: Resveratrol induction of TGF-beta expression and signaling. FEBS Lett. 2008, 582, 586–590. [Google Scholar] [CrossRef] [Green Version]
  139. Garcia, P.; Schmiedlin-Ren, P.; Mathias, J.S.; Tang, H.; Christman, G.M.; Zimmermann, E.M. Resveratrol causes cell cycle arrest, decreased collagen synthesis, and apoptosis in rat intestinal smooth muscle cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 302, G326–G335. [Google Scholar] [CrossRef] [Green Version]
  140. Trotta, V.; Lee, W.H.; Loo, C.Y.; Haghi, M.; Young, P.M.; Scalia, S.; Traini, D. In vitro biological activity of resveratrol using a novel inhalable resveratrol spray-dried formulation. Int. J. Pharm. 2015, 491, 190–197. [Google Scholar] [CrossRef]
  141. Rahal, K.; Schmiedlin-Ren, P.; Adler, J.; Dhanani, M.; Sultani, V.; Rittershaus, A.C.; Reingold, L.; Zhu, J.; McKenna, B.J.; Christman, G.M.; et al. Resveratrol has antiinflammatory and antifibrotic effects in the peptidoglycan-polysaccharide rat model of Crohn’s disease. Inflamm. Bowel Dis. 2012, 18, 613–623. [Google Scholar] [CrossRef] [Green Version]
  142. Wei, G.; Chen, X.; Wang, G.; Fan, L.; Wang, K.; Li, X. Effect of Resveratrol on the Prevention of Intra-Abdominal Adhesion Formation in a Rat Model. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2016, 39, 33–46. [Google Scholar] [CrossRef]
  143. He, Y.; Zeng, H.; Yu, Y.; Zhang, J.; Liu, Q.; Yang, B. Resveratrol improved detrusor fibrosis induced by mast cells during progression of chronic prostatitis in rats. Eur. J. Pharmacol. 2017, 815, 495–500. [Google Scholar] [CrossRef]
  144. Alrafas, H.R.; Busbee, P.B.; Nagarkatti, M.; Nagarkatti, P.S. Resveratrol Downregulates miR-31 to Promote T Regulatory Cells during Prevention of TNBS-Induced Colitis. Mol. Nutr. Food Res. 2020, 64, e1900633. [Google Scholar] [CrossRef]
  145. Xiao, Z.; Chen, C.; Meng, T.; Zhang, W.; Zhou, Q. Resveratrol attenuates renal injury and fibrosis by inhibiting transforming growth factor-β pathway on matrix metalloproteinase 7. Exp. Biol. Med. 2016, 241, 140–146. [Google Scholar] [CrossRef] [Green Version]
  146. Ishikawa, K.; He, S.; Terasaki, H.; Nazari, H.; Zhang, H.; Spee, C.; Kannan, R.; Hinton, D.R. Resveratrol inhibits epithelial-mesenchymal transition of retinal pigment epithelium and development of proliferative vitreoretinopathy. Sci. Rep. 2015, 5, 16386. [Google Scholar] [CrossRef]
  147. Rosa, P.M.; Martins, L.A.M.; Souza, D.O.; Quincozes-Santos, A. Glioprotective Effect of Resveratrol: An Emerging Therapeutic Role for Oligodendroglial Cells. Mol. Neurobiol. 2018, 55, 2967–2978. [Google Scholar] [CrossRef]
  148. Losso, J.N.; Truax, R.E.; Richard, G. trans-resveratrol inhibits hyperglycemia-induced inflammation and connexin downregulation in retinal pigment epithelial cells. J. Agric. Food Chem 2010, 58, 8246–8252. [Google Scholar] [CrossRef]
  149. Das, S.K.; Mukherjee, S.; Gupta, G.; Rao, D.N.; Vasudevan, D.M. Protective effect of resveratrol and vitamin E against ethanol-induced oxidative damage in mice: Biochemical and immunological basis. Indian J. Biochem. Biophys. 2010, 47, 32–37. [Google Scholar]
  150. Leppäranta, O.; Sens, C.; Salmenkivi, K.; Kinnula, V.L.; Keski-Oja, J.; Myllärniemi, M.; Koli, K. Regulation of TGF-β storage and activation in the human idiopathic pulmonary fibrosis lung. Cell Tissue Res. 2012, 348, 491–503. [Google Scholar] [CrossRef]
  151. Bellaye, P.S.; Yanagihara, T.; Granton, E.; Sato, S.; Shimbori, C.; Upagupta, C.; Imani, J.; Hambly, N.; Ask, K.; Gauldie, J.; et al. Macitentan reduces progression of TGF-β1-induced pulmonary fibrosis and pulmonary hypertension. Eur. Respir. J. 2018, 52. [Google Scholar] [CrossRef]
  152. Wang, J.; He, F.; Chen, L.; Li, Q.; Jin, S.; Zheng, H.; Lin, J.; Zhang, H.; Ma, S.; Mei, J.; et al. Resveratrol inhibits pulmonary fibrosis by regulating miR-21 through MAPK/AP-1 pathways. Biomed. Pharmacother. 2018, 105, 37–44. [Google Scholar] [CrossRef]
  153. Gao, C.; Howard-Quijano, K.; Rau, C.; Takamiya, T.; Song, Y.; Shivkumar, K.; Wang, Y.; Mahajan, A. Inflammatory and apoptotic remodeling in autonomic nervous system following myocardial infarction. PLoS ONE 2017, 12, e0177750. [Google Scholar] [CrossRef]
  154. Gao, H.; Bo, Z.; Wang, Q.; Luo, L.; Zhu, H.; Ren, Y. Salvanic acid B inhibits myocardial fibrosis through regulating TGF-β1/Smad signaling pathway. Biomed. Pharmacother. 2019, 110, 685–691. [Google Scholar] [CrossRef]
  155. Zhang, Y.; Lu, Y.; Ong’achwa, M.J.; Ge, L.; Qian, Y.; Chen, L.; Hu, X.; 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]
  156. Annaldas, S.; Saifi, M.A.; Khurana, A.; Godugu, C. Nimbolide ameliorates unilateral ureteral obstruction-induced renal fibrosis by inhibition of TGF-β and EMT/Slug signalling. Mol. Immunol. 2019, 112, 247–255. [Google Scholar] [CrossRef]
  157. Song, M.K.; Lee, J.H.; Ryoo, I.G.; Lee, S.H.; Ku, S.K.; Kwak, M.K. Bardoxolone ameliorates TGF-β1-associated renal fibrosis through Nrf2/Smad7 elevation. Free Radic. Biol. Med. 2019, 138, 33–42. [Google Scholar] [CrossRef]
  158. Zhang, X.; Lu, H.; Xie, S.; Wu, C.; Guo, Y.; Xiao, Y.; Zheng, S.; Zhu, H.; Zhang, Y.; Bai, Y. Resveratrol suppresses the myofibroblastic phenotype and fibrosis formation in kidneys via proliferation-related signalling pathways. Br. J. Pharmacol. 2019, 176, 4745–4759. [Google Scholar] [CrossRef]
  159. Liu, Y. New insights into epithelial-mesenchymal transition in kidney fibrosis. J. Am. Soc. Nephrol. 2010, 21, 212–222. [Google Scholar] [CrossRef] [Green Version]
  160. Alpers, C.E.; Hudkins, K.L.; Floege, J.; Johnson, R.J. Human renal cortical interstitial cells with some features of smooth muscle cells participate in tubulointerstitial and crescentic glomerular injury. J. Am. Soc. Nephrol. 1994, 5, 201–209. [Google Scholar]
  161. Bi, W.; Xu, G.; Lv, L.; Yang, C. The ratio of transforming growth factor-β1/bone morphogenetic protein-7 in the progression of the epithelial-mesenchymal transition contributes to rat liver fibrosis. Genet. Mol. Res. 2014, 13, 1005–1014. [Google Scholar] [CrossRef]
  162. Kalluri, R.; Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Investig. 2009, 119, 1420–1428. [Google Scholar] [CrossRef] [Green Version]
  163. Bai, Y.; Lu, H.; Wu, C.; Liang, Y.; Wang, S.; Lin, C.; Chen, B.; Xia, P. Resveratrol inhibits epithelial-mesenchymal transition and renal fibrosis by antagonizing the hedgehog signaling pathway. Biochem. Pharmacol. 2014, 92, 484–493. [Google Scholar] [CrossRef]
  164. Zhai, X.X.; Ding, J.C.; Tang, Z.M. Resveratrol Inhibits Proliferation and Induces Apoptosis of Pathological Scar Fibroblasts Through the Mechanism Involving TGF-β1/Smads Signaling Pathway. Cell Biochem. Biophys. 2015, 71, 1267–1272. [Google Scholar] [CrossRef]
  165. Liu, S.; Zhao, M.; Zhou, Y.; Wang, C.; Yuan, Y.; Li, L.; Bresette, W.; Chen, Y.; Cheng, J.; Lu, Y.; et al. Resveratrol exerts dose-dependent anti-fibrotic or pro-fibrotic effects in kidneys: A potential risk to individuals with impaired kidney function. Phytomedicine 2019, 57, 223–235. [Google Scholar] [CrossRef]
  166. Chávez, E.; Reyes-Gordillo, K.; Segovia, J.; Shibayama, M.; Tsutsumi, V.; Vergara, P.; Moreno, M.G.; Muriel, P. Resveratrol prevents fibrosis, NF-kappaB activation and TGF-beta increases induced by chronic CCl4 treatment in rats. J. Appl. Toxicol. JAT 2008, 28, 35–43. [Google Scholar] [CrossRef]
  167. Ding, S.; Wang, H.; Wang, M.; Bai, L.; Yu, P.; Wu, W. Resveratrol alleviates chronic “real-world” ambient particulate matter-induced lung inflammation and fibrosis by inhibiting NLRP3 inflammasome activation in mice. Ecotoxicol. Environ. Saf. 2019, 182, 109425. [Google Scholar] [CrossRef]
  168. Sun, D.Y.; Wu, J.Q.; He, Z.H.; He, M.F.; Sun, H.B. Cancer-associated fibroblast regulate proliferation and migration of prostate cancer cells through TGF-β signaling pathway. Life Sci. 2019, 235, 116791. [Google Scholar] [CrossRef]
  169. Cruz-Bermúdez, A.; Laza-Briviesca, R.; Vicente-Blanco, R.J.; García-Grande, A.; Coronado, M.J.; Laine-Menéndez, S.; Alfaro, C.; Sanchez, J.C.; Franco, F.; Calvo, V.; et al. Cancer-associated fibroblasts modify lung cancer metabolism involving ROS and TGF-β signaling. Free Radic. Biol. Med. 2019, 130, 163–173. [Google Scholar] [CrossRef]
  170. Bierie, B.; Moses, H.L. TGF-beta and cancer. Cytokine Growth Factor Rev. 2006, 17, 29–40. [Google Scholar] [CrossRef]
  171. Zhao, M.; Mishra, L.; Deng, C.X. The role of TGF-β/SMAD4 signaling in cancer. Int. J. Biol. Sci. 2018, 14, 111–123. [Google Scholar] [CrossRef] [Green Version]
  172. Camerlingo, R.; Miceli, R.; Marra, L.; Rea, G.; D’Agnano, I.; Nardella, M.; Montella, R.; Morabito, A.; Normanno, N.; Tirino, V.; et al. Conditioned medium of primary lung cancer cells induces EMT in A549 lung cancer cell line by TGF-ß1 and miRNA21 cooperation. PLoS ONE 2019, 14, e0219597. [Google Scholar] [CrossRef] [Green Version]
  173. Liu, X.S.; Lin, X.K.; Mei, Y.; Ahmad, S.; Yan, C.X.; Jin, H.L.; Yu, H.; Chen, C.; Lin, C.Z.; Yu, J.R. Regulatory T Cells Promote Overexpression of Lgr5 on Gastric Cancer Cells via TGF-beta1 and Confer Poor Prognosis in Gastric Cancer. Front. Immunol. 2019, 10, 1741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Hyun Lee, J.; Dhananjaya Mohan, C.; Deivasigamani, A.; Yun Jung, Y.; Rangappa, S.; Basappa, S.; Chinnathambi, A.; Awad Alahmadi, T.; Ali Alharbi, S.; Garg, M.; et al. Brusatol suppresses STAT3-driven metastasis by downregulating epithelial-mesenchymal transition in hepatocellular carcinoma. J. Adv. Res. 2020. [Google Scholar] [CrossRef]
  175. Cheng, J.-T.; Wang, L.; Wang, H.; Tang, F.-R.; Cai, W.-Q.; Sethi, G.; Xin, H.-W.; Ma, Z. Insights into Biological Role of LncRNAs in Epithelial-Mesenchymal Transition. Cells 2019, 8, 1178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Loh, C.-Y.; Chai, J.Y.; Tang, T.F.; Wong, W.F.; Sethi, G.; Shanmugam, M.K.; Chong, P.P.; Looi, C.Y. The E-Cadherin and N-Cadherin Switch in Epithelial-to-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells 2019, 8, 1118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Sun, Y.; Zhou, Q.M.; Lu, Y.Y.; Zhang, H.; Chen, Q.L.; Zhao, M.; Su, S.B. 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]
  178. Wang, H.; Zhang, H.; Tang, L.; Chen, H.; Wu, C.; Zhao, M.; Yang, Y.; Chen, X.; Liu, G. Resveratrol inhibits TGF-β1-induced epithelial-to-mesenchymal transition and suppresses lung cancer invasion and metastasis. Toxicology 2013, 303, 139–146. [Google Scholar] [CrossRef]
  179. 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]
  180. 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]
  181. Zhang, Y.; Yang, S.; Yang, Y.; Liu, T. Resveratrol induces immunogenic cell death of human and murine ovarian carcinoma cells. Infect. Agents Cancer 2019, 14, 27. [Google Scholar] [CrossRef] [Green Version]
  182. Rekik, R.; Belhadj Hmida, N.; Ben Hmid, A.; Zamali, I.; Kammoun, N.; Ben Ahmed, M. PD-1 induction through TCR activation is partially regulated by endogenous TGF-β. Cell. Mol. Immunol. 2015, 12, 648–649. [Google Scholar] [CrossRef] [Green Version]
  183. Celada, L.J.; Kropski, J.A.; Herazo-Maya, J.D.; Luo, W.; Creecy, A.; Abad, A.T.; Chioma, O.S.; Lee, G.; Hassell, N.E.; Shaginurova, G.I.; et al. PD-1 up-regulation on CD4(+) T cells promotes pulmonary fibrosis through STAT3-mediated IL-17A and TGF-β1 production. Sci. Transl. Med. 2018, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Johnson, E.R.; Matthay, M.A. Acute lung injury: Epidemiology, pathogenesis, and treatment. J. Aerosol Med. Pulm. Drug Deliv. 2010, 23, 243–252. [Google Scholar] [CrossRef] [PubMed]
  185. Sawa, T. The molecular mechanism of acute lung injury caused by Pseudomonas aeruginosa: From bacterial pathogenesis to host response. J. Intensive Care 2014, 2, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Kubota, Y.; Iwasaki, Y.; Harada, H.; Yokomura, I.; Ueda, M.; Hashimoto, S.; Nakagawa, M. Role of alveolar macrophages in Candida-induced acute lung injury. Clin. Diagn. Lab. Immunol. 2001, 8, 1258–1262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Savransky, V.; Rostapshov, V.; Pinelis, D.; Polotsky, Y.; Korolev, S.; Komisar, J.; Fegeding, K. Murine lethal toxic shock caused by intranasal administration of staphylococcal enterotoxin B. Toxicol. Pathol. 2003, 31, 373–378. [Google Scholar] [CrossRef] [PubMed]
  188. 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]
  189. Karagiannidis, C.; Akdis, M.; Holopainen, P.; Woolley, N.J.; Hense, G.; Rückert, B.; Mantel, P.Y.; Menz, G.; Akdis, C.A.; Blaser, K.; et al. Glucocorticoids upregulate FOXP3 expression and regulatory T cells in asthma. J. Allergy Clin. Immunol. 2004, 114, 1425–1433. [Google Scholar] [CrossRef]
  190. Alharris, E.; Alghetaa, H.; Seth, R.; Chatterjee, S.; Singh, N.P.; Nagarkatti, M.; Nagarkatti, P. Resveratrol Attenuates Allergic Asthma and Associated Inflammation in the Lungs Through Regulation of miRNA-34a That Targets FoxP3 in Mice. Front. Immunol. 2018, 9, 2992. [Google Scholar] [CrossRef] [Green Version]
  191. Wollin, L.; Pieper, M. Tiotropium bromide exerts anti-inflammatory activity in a cigarette smoke mouse model of COPD. Pulm. Pharmacol. Ther. 2010, 23, 345–354. [Google Scholar] [CrossRef]
  192. Tamimi, A.; Serdarevic, D.; Hanania, N.A. The effects of cigarette smoke on airway inflammation in asthma and COPD: Therapeutic implications. Respir. Med. 2012, 106, 319–328. [Google Scholar] [CrossRef] [Green Version]
  193. Busse, P.J.; Zhang, T.F.; Srivastava, K.; Lin, B.P.; Schofield, B.; Sealfon, S.C.; Li, X.-M. Chronic exposure to TNF-α increases airway mucus gene expression in vivo. J. Allergy Clin. Immunol. 2005, 116, 1256–1263. [Google Scholar] [CrossRef] [PubMed]
  194. Numasaki, M.; Tomioka, Y.; Takahashi, H.; Sasaki, H. IL-17 and IL-17F modulate GM-CSF production by lung microvascular endothelial cells stimulated with IL-1β and/or TNF-α. Immunol. Lett. 2004, 95, 175–184. [Google Scholar] [CrossRef] [PubMed]
  195. Chen, J.; Yang, X.; Zhang, W.; Peng, D.; Xia, Y.; Lu, Y.; Han, X.; Song, G.; Zhu, J.; Liu, R. Therapeutic Effects of Resveratrol in a Mouse Model of LPS and Cigarette Smoke-Induced COPD. Inflammation 2016, 39, 1949–1959. [Google Scholar] [CrossRef] [PubMed]
  196. Al-Mufti, F.; Amuluru, K.; Changa, A.; Lander, M.; Patel, N.; Wajswol, E.; Al-Marsoummi, S.; Alzubaidi, B.; Singh, I.P.; Nuoman, R.; et al. Traumatic brain injury and intracranial hemorrhage-induced cerebral vasospasm: A systematic review. Neurosurg. Focus 2017, 43, E14. [Google Scholar] [CrossRef] [Green Version]
  197. Shoamanesh, A.; Kwok, C.S.; Lim, P.A.; Benavente, O.R. Postthrombolysis intracranial hemorrhage risk of cerebral microbleeds in acute stroke patients: A systematic review and meta-analysis. Int. J. Stroke Off. J. Int. Stroke Soc. 2013, 8, 348–356. [Google Scholar] [CrossRef] [Green Version]
  198. Bugeme, M.; Mukuku, O. Neuropsychiatric manifestations revealing cerebral subarachnoid hemorrhage caused by electrification accident about a case and review of literature. Pan Afr. Med. J. 2014, 18, 201. [Google Scholar] [CrossRef]
  199. Logan, T.T.; Villapol, S.; Symes, A.J. TGF-β superfamily gene expression and induction of the Runx1 transcription factor in adult neurogenic regions after brain injury. PLoS ONE 2013, 8, e59250. [Google Scholar] [CrossRef] [Green Version]
  200. Zhao, R.; Zhao, K.; Su, H.; Zhang, P.; Zhao, N. Resveratrol ameliorates brain injury via the TGF-β-mediated ERK signaling pathway in a rat model of cerebral hemorrhage. Exp. Ther. Med. 2019, 18, 3397–3404. [Google Scholar] [CrossRef] [Green Version]
  201. Tiwari, V.; Chopra, K. Resveratrol prevents alcohol-induced cognitive deficits and brain damage by blocking inflammatory signaling and cell death cascade in neonatal rat brain. J. Neurochem. 2011, 117, 678–690. [Google Scholar] [CrossRef]
  202. Forbes, J.M.; Cooper, M.E.; Oldfield, M.D.; Thomas, M.C. Role of advanced glycation end products in diabetic nephropathy. J. Am. Soc. Nephrol. 2003, 14, S254–S258. [Google Scholar] [CrossRef] [Green Version]
  203. Yamagishi, S.-i.; Matsui, T. Advanced glycation end products, oxidative stress and diabetic nephropathy. Oxidative Med. Cell. Longev. 2010, 3, 101–108. [Google Scholar] [CrossRef] [PubMed]
  204. Mima, A. Inflammation and oxidative stress in diabetic nephropathy: New insights on its inhibition as new therapeutic targets. J. Diabetes Res. 2013, 2013, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Hussein, M.M.; Mahfouz, M.K. Effect of resveratrol and rosuvastatin on experimental diabetic nephropathy in rats. Biomed. Pharmacother. 2016, 82, 685–692. [Google Scholar] [CrossRef] [PubMed]
  206. Chen, K.H.; Hung, C.C.; Hsu, H.H.; Jing, Y.H.; Yang, C.W.; Chen, J.K. Resveratrol ameliorates early diabetic nephropathy associated with suppression of augmented TGF-β/smad and ERK1/2 signaling in streptozotocin-induced diabetic rats. Chem. Biol. Interact. 2011, 190, 45–53. [Google Scholar] [CrossRef] [PubMed]
  207. Abd El-Haleim, E.A.; Bahgat, A.K.; Saleh, S. Resveratrol and fenofibrate ameliorate fructose-induced nonalcoholic steatohepatitis by modulation of genes expression. World J. Gastroenterol. 2016, 22, 2931–2948. [Google Scholar] [CrossRef]
  208. Qiao, Y.; Gao, K.; Wang, Y.; Wang, X.; Cui, B. Resveratrol ameliorates diabetic nephropathy in rats through negative regulation of the p38 MAPK/TGF-β1 pathway. Exp. Ther. Med. 2017, 13, 3223–3230. [Google Scholar] [CrossRef] [Green Version]
  209. Wu, H.; Li, G.N.; Xie, J.; Li, R.; Chen, Q.H.; Chen, J.Z.; Wei, Z.H.; Kang, L.N.; Xu, B. Resveratrol ameliorates myocardial fibrosis by inhibiting ROS/ERK/TGF-β/periostin pathway in STZ-induced diabetic mice. BMC Cardiovasc. Disord. 2016, 16, 5. [Google Scholar] [CrossRef] [Green Version]
  210. Elbe, H.; Vardi, N.; Esrefoglu, M.; Ates, B.; Yologlu, S.; Taskapan, C. Amelioration of streptozotocin-induced diabetic nephropathy by melatonin, quercetin, and resveratrol in rats. Hum. Exp. Toxicol. 2015, 34, 100–113. [Google Scholar] [CrossRef]
  211. Wenbin, Z.; Guojun, G. Resveratrol Ameliorates Diabetes-induced Renal Damage through Regulating the Expression of TGF-β1, Collagen IV and Th17/Treg-related Cytokines in Rats. West Indian Med. J. 2014, 63, 20–25. [Google Scholar] [CrossRef] [Green Version]
  212. Liu, J.; Zhuo, X.; Liu, W.; Wan, Z.; Liang, X.; Gao, S.; Yuan, Z.; Wu, Y. Resveratrol inhibits high glucose induced collagen upregulation in cardiac fibroblasts through regulating TGF-β1-Smad3 signaling pathway. Chem. Biol. Interact. 2015, 227, 45–52. [Google Scholar] [CrossRef]
  213. Hammad, A.S.A.; Ahmed, A.F.; Heeba, G.H.; Taye, A. Heme oxygenase-1 contributes to the protective effect of resveratrol against endothelial dysfunction in STZ-induced diabetes in rats. Life Sci. 2019, 239, 117065. [Google Scholar] [CrossRef] [PubMed]
  214. Zeng, H.; He, Y.; Yu, Y.; Zhang, J.; Zeng, X.; Gong, F.; Liu, Q.; Yang, B. Resveratrol improves prostate fibrosis during progression of urinary dysfunction in chronic prostatitis by mast cell suppression. Mol. Med. Rep. 2018, 17, 918–924. [Google Scholar] [CrossRef] [PubMed]
  215. He, Y.; Zeng, H.Z.; Yu, Y.; Zhang, J.S.; Duan, X.; Zeng, X.N.; Gong, F.T.; Liu, Q.; Yang, B. Resveratrol improves prostate fibrosis during progression of urinary dysfunction in chronic prostatitis. Environ. Toxicol. Pharmacol. 2017, 54, 120–124. [Google Scholar] [CrossRef] [PubMed]
  216. Zhang, Y.Q.; Liu, Y.J.; Mao, Y.F.; Dong, W.W.; Zhu, X.Y.; Jiang, L. Resveratrol ameliorates lipopolysaccharide-induced epithelial mesenchymal transition and pulmonary fibrosis through suppression of oxidative stress and transforming growth factor-β1 signaling. Clin. Nutr. 2015, 34, 752–760. [Google Scholar] [CrossRef] [PubMed]
  217. Shi, X.P.; Miao, S.; Wu, Y.; Zhang, W.; Zhang, X.F.; Ma, H.Z.; Xin, H.L.; Feng, J.; Wen, A.D.; Li, Y. Resveratrol sensitizes tamoxifen in antiestrogen-resistant breast cancer cells with epithelial-mesenchymal transition features. Int. J. Mol. Sci. 2013, 14, 15655–15668. [Google Scholar] [CrossRef] [PubMed]
  218. Kim, K.H.; Back, J.H.; Zhu, Y.; Arbesman, J.; Athar, M.; Kopelovich, L.; Kim, A.L.; Bickers, D.R. Resveratrol targets transforming growth factor-β2 signaling to block UV-induced tumor progression. J. Investig. Dermatol. 2011, 131, 195–202. [Google Scholar] [CrossRef] [Green Version]
  219. Kim, J.S.; Jeong, S.K.; Oh, S.J.; Lee, C.G.; Kang, Y.R.; Jo, W.S.; Jeong, M.H. The resveratrol analogue, HS-1793, enhances the effects of radiation therapy through the induction of anti-tumor immunity in mammary tumor growth. Int. J. Oncol. 2020, 56, 1405–1416. [Google Scholar] [CrossRef] [Green Version]
  220. Chen, T.; Li, J.; Liu, J.; Li, N.; Wang, S.; Liu, H.; Zeng, M.; Zhang, Y.; Bu, P. Activation of SIRT3 by resveratrol ameliorates cardiac fibrosis and improves cardiac function via the TGF-β/Smad3 pathway. Am. J. Physiol. Heart Circ. Physiol. 2015, 308, H424–H434. [Google Scholar] [CrossRef]
  221. Lee, H.Y.; Kim, I.K.; Yoon, H.K.; Kwon, S.S.; Rhee, C.K.; Lee, S.Y. Inhibitory Effects of Resveratrol on Airway Remodeling by Transforming Growth Factor-β/Smad Signaling Pathway in Chronic Asthma Model. Allergyasthma Immunol. Res. 2017, 9, 25–34. [Google Scholar] [CrossRef] [Green Version]
  222. Chen, C.L.; Chen, Y.H.; Tai, M.C.; Liang, C.M.; Lu, D.W.; Chen, J.T. Resveratrol inhibits transforming growth factor-β2-induced epithelial-to-mesenchymal transition in human retinal pigment epithelial cells by suppressing the Smad pathway. Drug Des. Dev. Ther. 2017, 11, 163–173. [Google Scholar] [CrossRef] [Green Version]
  223. Hori, Y.S.; Kuno, A.; Hosoda, R.; Tanno, M.; Miura, T.; Shimamoto, K.; Horio, Y. Resveratrol ameliorates muscular pathology in the dystrophic mdx mouse, a model for Duchenne muscular dystrophy. J. Pharmacol. Exp. Ther. 2011, 338, 784–794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Yang, H.; Yuan, Y.; Luo, C.; He, H.; Zhou, Y. Inhibitory Effects of Resveratrol on the Human Alveolar Rhabdomyosarcoma Cell Line PLA-802 through Inhibition of the TGF-β1/Smad Signaling Pathway. Pharmacology 2016, 98, 35–41. [Google Scholar] [CrossRef] [PubMed]
  225. Li, J.; Qu, X.; Ricardo, S.D.; Bertram, J.F.; Nikolic-Paterson, D.J. Resveratrol inhibits renal fibrosis in the obstructed kidney: Potential role in deacetylation of Smad3. Am. J. Pathol. 2010, 177, 1065–1071. [Google Scholar] [CrossRef] [PubMed]
  226. Liskova, A.; Koklesova, L.; Samec, M.; Smejkal, K.; Samuel, S.M.; Varghese, E.; Abotaleb, M.; Biringer, K.; Kudela, E.; Danko, J.; et al. Flavonoids in Cancer Metastasis. Cancers 2020, 12, 1498. [Google Scholar] [CrossRef] [PubMed]
  227. Manu, K.A.; Shanmugam, M.K.; Ramachandran, L.; Li, F.; Siveen, K.S.; Chinnathambi, A.; Zayed, M.E.; Alharbi, S.A.; Arfuso, F.; Kumar, A.P.; et al. Isorhamnetin augments the anti-tumor effect of capeciatbine through the negative regulation of NF-κB signaling cascade in gastric cancer. Cancer Lett. 2015, 363, 28–36. [Google Scholar] [CrossRef]
  228. Varghese, E.; Liskova, A.; Kubatka, P.; Samuel, S.M.; Büsselberg, D. Anti-Angiogenic Effects of Phytochemicals on miRNA Regulating Breast Cancer Progression. Biomolecules 2020, 10, 191. [Google Scholar] [CrossRef] [Green Version]
  229. Manu, K.A.; Shanmugam, M.K.; Li, F.; Chen, L.; Siveen, K.S.; Ahn, K.S.; Kumar, A.P.; Sethi, G. Simvastatin sensitizes human gastric cancer xenograft in nude mice to capecitabine by suppressing nuclear factor-kappa B-regulated gene products. J. Mol. Med. 2014, 92, 267–276. [Google Scholar] [CrossRef] [Green Version]
  230. Siveen, K.S.; Ahn, K.S.; Ong, T.H.; Shanmugam, M.K.; Li, F.; Yap, W.N.; Kumar, A.P.; Fong, C.W.; Tergaonkar, V.; Hui, K.M.; et al. γ-tocotrienol inhibits angiogenesis-dependent growth of human hepatocellular carcinoma through abrogation of AKT/mTOR pathway in an orthotopic mouse model. Oncotarget 2014, 5, 1897–1911. [Google Scholar] [CrossRef] [Green Version]
  231. Shanmugam, M.K.; Kannaiyan, R.; Sethi, G. Targeting cell signaling and apoptotic pathways by dietary agents: Role in the prevention and treatment of cancer. Nutr. Cancer 2011, 63, 161–173. [Google Scholar] [CrossRef]
  232. Sawhney, M.; Rohatgi, N.; Kaur, J.; Shishodia, S.; Sethi, G.; Gupta, S.D.; Deo, S.V.; Shukla, N.K.; Aggarwal, B.B.; Ralhan, R. Expression of NF-kappaB parallels COX-2 expression in oral precancer and cancer: Association with smokeless tobacco. Int. J. Cancer 2007, 120, 2545–2556. [Google Scholar] [CrossRef]
  233. Ahn, K.S.; Sethi, G.; Jain, A.K.; Jaiswal, A.K.; Aggarwal, B.B. Genetic deletion of NAD(P)H:quinone oxidoreductase 1 abrogates activation of nuclear factor-kappaB, IkappaBalpha kinase, c-Jun N-terminal kinase, Akt, p38, and p44/42 mitogen-activated protein kinases and potentiates apoptosis. J. Biol. Chem. 2006, 281, 19798–19808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. A schematic presentation of transforming growth factor-beta (TGF-β) signaling pathways. This pathway consists of two distinct modules: canonical signaling and non-canonical signaling. Canonical signaling, as shown in the figure, is a result of the formation of a complex containing Smad2, Smad3, and Smad4. Then, these molecules can translocate into the nucleus to trigger the expression of genes that are responsible for the proliferation and metastasis of cancer cells. Non-canonical signaling is Smad-independent and involves different signaling pathways such as PI3K/Akt, MAP3K7, Ras, and so on. However, final aim of these two signaling pathways is to promote aberrant growth and malignancy of cancer cells.
Figure 1. A schematic presentation of transforming growth factor-beta (TGF-β) signaling pathways. This pathway consists of two distinct modules: canonical signaling and non-canonical signaling. Canonical signaling, as shown in the figure, is a result of the formation of a complex containing Smad2, Smad3, and Smad4. Then, these molecules can translocate into the nucleus to trigger the expression of genes that are responsible for the proliferation and metastasis of cancer cells. Non-canonical signaling is Smad-independent and involves different signaling pathways such as PI3K/Akt, MAP3K7, Ras, and so on. However, final aim of these two signaling pathways is to promote aberrant growth and malignancy of cancer cells.
Biomedicines 08 00261 g001
Figure 2. Regulation of TGF-β signaling by Res and its association with therapeutic effects.
Figure 2. Regulation of TGF-β signaling by Res and its association with therapeutic effects.
Biomedicines 08 00261 g002
Table 1. Res targets TGF-β signaling pathway in disease therapy.
Table 1. Res targets TGF-β signaling pathway in disease therapy.
DrugIn Vitro/In VivoDiseaseDoseDuration of ExperimentAdministration RouteEffect on TGF-βResultsReferences
Resveratrol
Fenofibrate
In vivo (animal model of steatohepatitis)Steatohepatitis70 mg/kg12 weeksDietInhibitionAlleviation of nonalcoholic steatohepatitis[207]
ResveratrolIn vitro (rat mesangial cells)
In vivo (rat model of diabetic nephropathy)
Diabetes25 µM
20 mg/kg
24 h
4 weeks
OralInhibitionReducing mesangial cell viability, fibronectin secretion, and amelioration of diabetic nephropathy[208]
ResveratrolIn vivo (diabetic mice)Diabetes5 and 25 mg/kg/day2 monthsIntragastricInhibitionImproving fibrosis via inhibition of ROS/ERK/TGF-β[209]
ResveratrolIn vivo (diabetic rats)Diabetes10 mg/kg/day30 daysIntraperitonealInhibitionAlleviation of diabetic nephropathy and reducing epithelial desquamation, swelling, intracytoplasmic vacuolization, brush border loss, and peritubular infiltration[210]
ResveratrolIn vivo (diabetic rats)Diabetes50 mg/kg8 weeksGavageInhibitionAmelioration of renal damage and reducing collagen deposition[211]
ResveratrolIn vivo (diabetic model)Diabetes10 mg/kg8 weeksOral gavageInhibitionReducing collagen deposition[212]
ResveratrolIn vivo (diabetic rats)Diabetes10 mg/kg4 weeksDrinking waterInhibitionImproving vascular dysfunction and reducing oxidative stress[213]
ResveratrolIn vivo (rat model of chronic prostatitis)Chronic prostatitis10 mg/kg10 daysOralInhibitionAlleviation of prostate fibrosis via mast cell suppression[214]
ResveratrolIn vivo (rat model of chronic prostatitis)Chronic prostatitis10 mg/kg10 daysOralInhibitionReducing prostate fibrosis and urinary dysfunction via inhibition of TGF-β/Wnt/β-catenin[215]
ResveratrolIn vitro (Human colorectal cancer cell line LoVo)
In vivo (mice with orthotopic transplantation tumor)
Cancer6 and 12 µM
50, 100, and 150 mg/kg
24 h
3 weeks
IntragastricInhibitionSuppressing metastasis of cancer cells by EMT inhibition via down-regulation of TGF-β/Smad signaling pathway[216]
ResveratrolIn vitro (MCF-7 cells)Cancer5, 25, 50, 100, and 200 µM48 h-InhibitionSensitizing cancer cells into chemotherapy via inhibition of TGF-β-mediated EMT[217]
ResveratrolIn vitro (A431 human epidermoid carcinoma cells)Cancer50–100 µM24 h-InhibitionSuppressing ultraviolet-induced tumor proliferation[218]
Resveratrol analogue (HS-1793)In vivo (tumor bearing mice)Cancer0.5 and 1 mg/kg3 weeksIntraperitonealInhibitionEnhancing efficacy of radiotherapy[219]
ResveratrolMurine model of LPS-induced pulmonary fibrosisPulmonary fibrosis0.3 mg/kg28 daysIntraperitonealInhibitionImproving pulmonary fibrosis and inhibition of EMT via the down-regulation of TGF-β1/Smad[216]
ResveratrolIn vivo (SIRT3-knock out mice)Fibrosis1.8 mg/kg8 weeksDietInhibitionImproving cardiac fibrosis and suppressing fibroblast-to-myoblast transformation[220]
ResveratrolIn vivo (chronic asthma model)Asthma10 and 50 mg/kg3 monthsOral gavageInhibitionInhibition of Smad2/3 phosphorylation, amelioration of airway inflammation and structural changes[221]
ResveratrolIn vitro (human retinal pigment epithelial cells)Eye disease25, 50, 100, 200, 400, and 800 µM24 h-InhibitionSuppressing Smad2 and Smad3 phosphorylation leads to the inhibition of EMT and collagen deposition[222]
ResveratrolIn vivo (mouse model of Duchene muscular dystrophy)Muscular dystrophy4 g/kg32 weeksDietInhibitionDecreasing reactive oxygen species generation, fibronectin production, and enhancing expressions of α-SMA and SIRT1[223]
ResveratrolIn vitro (rhabdomyosarcoma)Rhabdomyosarcoma5, 10, 20, 40, or 80 μmol/L24, 48, and 72 h-InhibitionInduction of G1 and S phases cell cycle arrest and down-regulation of Smad4[224]
ResveratrolIn vivo (Male C57BL/6J mice)-5 mg/kg2 days after surgeryIntraperitonealInhibitionReducing levels of collagen IV and fibronectin[225]
TGF-β, transforming growth factor-beta; ROS, reactive oxygen species; ERK, extracellular signal-regulated kinase; EMT, epithelial-to-mesenchymal transition; α-SMA, α-smooth muscle actin, SIRT1, sirtuin 1.

Share and Cite

MDPI and ACS Style

Ashrafizadeh, M.; Najafi, M.; Orouei, S.; Zabolian, A.; Saleki, H.; Azami, N.; Sharifi, N.; Hushmandi, K.; Zarrabi, A.; Ahn, K.S. Resveratrol Modulates Transforming Growth Factor-Beta (TGF-β) Signaling Pathway for Disease Therapy: A New Insight into Its Pharmacological Activities. Biomedicines 2020, 8, 261. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines8080261

AMA Style

Ashrafizadeh M, Najafi M, Orouei S, Zabolian A, Saleki H, Azami N, Sharifi N, Hushmandi K, Zarrabi A, Ahn KS. Resveratrol Modulates Transforming Growth Factor-Beta (TGF-β) Signaling Pathway for Disease Therapy: A New Insight into Its Pharmacological Activities. Biomedicines. 2020; 8(8):261. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines8080261

Chicago/Turabian Style

Ashrafizadeh, Milad, Masoud Najafi, Sima Orouei, Amirhossein Zabolian, Hossein Saleki, Negar Azami, Negin Sharifi, Kiavash Hushmandi, Ali Zarrabi, and Kwang Seok Ahn. 2020. "Resveratrol Modulates Transforming Growth Factor-Beta (TGF-β) Signaling Pathway for Disease Therapy: A New Insight into Its Pharmacological Activities" Biomedicines 8, no. 8: 261. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines8080261

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