Next Article in Journal
Consumption of the Food Groups with the Revised Benefits in the New WIC Food Package: A Scoping Review
Previous Article in Journal
Patterns of Dietary Fatty Acids and Fat Spreads in Relation to Blood Pressure, Lipids and Insulin Resistance in Young Adults: A Repeat Cross-Sectional Study
Previous Article in Special Issue
Growth of Renal Cancer Cell Lines Is Strongly Inhibited by Synergistic Activity of Low-Dosed Amygdalin and Sulforaphane
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gnetin C in Cancer and Other Diseases: What Do We Know So Far?

by
Gisella Campanelli
and
Anait S. Levenson
*
Department of Veterinary Biomedical Sciences, Lewyt College of Veterinary Medicine, Long Island University, Brookville, NY 11548, USA
*
Author to whom correspondence should be addressed.
Nutrients 2025, 17(5), 863; https://doi.org/10.3390/nu17050863
Submission received: 4 February 2025 / Revised: 24 February 2025 / Accepted: 26 February 2025 / Published: 28 February 2025
(This article belongs to the Special Issue Anticancer Activities of Dietary Phytochemicals: 2nd Edition)

Abstract

:
Stilbenes are a class of natural polyphenols with multiple positive pharmacologic assets such as antioxidant, anti-inflammatory and anticancer effects. While monomeric stilbenes, represented mostly by resveratrol and pterostilbene, have been studied intensely in the last two decades, oligomeric compounds, which may have better prospects of becoming potent nutraceuticals, are much less studied. The goal of this review is to compile all available literature to date on the beneficial pharmacologic effects of Gnetin C, a resveratrol dimer, in cancer and other diseases. While studies have shown the beneficial effects of Gnetin C, as a single compound or a component of melinjo seed extract, through cellular models, in vivo preclinical studies are still lacking. This is except for prostate cancer, where various animal models, including xenografts and transgenic mice, have been used to evaluate Gnetin C’s more potent anti-inflammatory and anticancer effects compared to resveratrol and its monomeric analogs. Since Gnetin C’s safety has already been demonstrated in healthy volunteers, it is now logical to evaluate its efficacy for prostate cancer chemoprevention, interception and therapy in clinical trials.

1. Introduction

Natural polyphenols are plant-derived chemicals with beneficial effects on human health. Polyphenols are divided into different classes depending on their basic chemical structure. Stilbenes or stilbenoids contain two aromatic rings connected by a central ethylene double bond. Most stilbenes in plants are produced in response to environmental stress including ultraviolet radiation, injury or infection. One of the most well-known naturally occurring stilbenes is resveratrol (trans-3,4’,5-trihydroxystilbene), found in grapes and red wine. Resveratrol and pterostilbene (trans-3,5-dimethoxy-4’-hydroxystilbene), found mainly in blueberries, are by far the two most extensively studied stilbenes in terms of their beneficial bioactivity in different diseases. Both compounds have been shown to exert a range of biological activities including antifungal, antibacterial, antioxidative, anti-inflammatory, anticancer and cardio- and neuroprotective effects [1,2,3,4].
Plentiful information is available on the cellular and molecular mechanisms by which resveratrol and its monomeric analogs interfere with carcinogenesis, cancer progression, cancer resistance and metastasis. These stilbenes have been shown to affect multiple targets in signaling pathways associated with cell proliferation, inflammation, angiogenesis and metastatic potential. However, despite a large amount of in vitro and in vivo experimental data, neither resveratrol nor pterostilbene is used therapeutically because clinical trials have been inconclusive [5]. Among the reasons for resveratrol’s lack of success is its poor bioavailability due to rapid and extensive conjugation in the intestinal tract, resulting in metabolites that are less biologically active than the parent compound [6,7]. On the other hand, resveratrol’s methoxylated analog pterostilbene has greater anticancer activity due to its favorable pharmacokinetic parameters, with a bioavailability greater than ten times that of resveratrol [8].
In more recent years, various resveratrol oligomer derivatives have been described. These can occur either naturally or synthetically from the endogenous polymerization of two to eight resveratrol units to form dimers or more complex oligomers [9,10]. In many instances, these oligomers were found to exhibit superior beneficial activities compared to monomer derivatives [11,12,13,14]. The resveratrol dimer consisting of two trans-resveratrol units linked via a benzofuran ring, known as Gnetin C, is the focus of the current review (Figure 1). We will summarize what is known so far about the biological activities and molecular effects of Gnetin C in cancer and other human diseases.

2. Gnetin C, a Resveratrol Dimer

Gnetin C was isolated from the roots of the melinjo plant (Gnetum gnemon) belonging to the Gnetaceae family [15]. The seeds of melinjo also contain various resveratrol dimers including Gnetin C, gnemonoside A and gnemonoside D, as well as minor amounts of resveratrol [16]. Many of the health-promoting benefits of melinjo seed extract (MSE) are attributed to Gnetin C and its related substances.
Plants from the Gnetaceae family have long been used in traditional medicine for the treatment of bronchitis, arthritis, diabetes, heart problems and other diseases. The melinjo plant is native to rainforests in Malaysia, the Philippines, Singapore, northern India and French Polynesia. Its fruit, leaves and seeds are used in Indonesian cuisine. Crackers made from the seeds are the most commercially popular, while fruits and leaves are used in making soup or consumed as vegetables. Scientifically, Gnetin C, in either the purified form or in ethanolic extract mixtures, showed no toxicity in normal cells in culture [17,18,19] and in rodent in vivo studies [17,20]. Importantly, both MSE and Gnetin C appear to be safe in humans according to recent clinical trials [21,22,23,24,25].
Until recently, access to pure compounds was limited to their isolation from natural sources, which hampered their biological evaluation. Furthermore, the fraction of Gnetin C present in ethanolic extracts from melinjo plant can vary depending on the part of the plant and method of isolation used. While evaluation of such mixtures provides useful information, the specific effects contributed by Gnetin C alone are unclear. In the current review, we primarily discuss studies with purified Gnetin C used as a single agent compared to either ethanolic extracts from the melinjo plant or other purified stilbenes (monomers and dimers).

3. Gnetin C and Cancer

Cancer is a leading cause of morbidity and mortality. The safety of standard chemotherapeutic drugs is a concern, and consequently, natural polyphenols, among them stilbenes, have become the subject of increasing scientific interest for cancer prevention, interception and treatment.
Naturally occurring resveratrol oligomers, including Gnetin C, have been proposed as potential cancer chemopreventive compounds [26]. Summarized in Table 1 are the reported in vitro and in vivo studies evaluating the efficacy of Gnetin C compared to other stilbenes or MSE in preventing the development and progression of cancer through different signaling pathways.
In an acute myeloid leukemia (AML)-MT xenograft model, Gnetin C significantly lowered the development of leukemia and tumor incidence in the blood, bone marrow and spleen. Moreover, mechanistic studies using several AML and chronic myeloid leukemia (CML) cell lines showed the induction of cell cycle arrest and inhibition of ERK1/2 and AKT/mTOR pathways [18] (Table 1). It was also demonstrated that Gnetin C inhibited the growth of human leukemia HL60 cells with an IC50 value of 13 µM [29]. Inhibition of tumor angiogenesis was also demonstrated after treatment with 5% MSE. Furthermore, Gnetin C in comparison with resveratrol, other dimers and MSE in human umbilical vein endothelial cells (HUVECs) was found to have a superior inhibitory effect on VEGF- and BFGF-stimulated tube formation and ERK1/2-mediated cell proliferation and migration [19] (Table 1). Narayanan et al. evaluated Gnetin C anticancer effects in comparison with resveratrol and MSE against various cancer cell lines in vitro and demonstrated that Gnetin C was more effective in causing kinase 3/7-mediated apoptosis in these cells. In addition, in a colon-26 tumor-bearing mouse model, MSE reduced tumor growth, improved histology and inhibited angiogenesis and liver metastasis [17] (Table 1). Superior biological activity of Gnetin C compared to resveratrol was also shown in melanoma cells in vitro [27] (Table 1). Interestingly, in a separate study, pterostilbene-induced inhibition of VCAM-1 and Bcl-2 in B16-F10 metastatic melanoma cells reduced metastatic growth in the liver by decreasing the colonization ability of these cells [30]. Lastly, Gnetin C in comparison with resveratrol and MSE showed more potent immunomodulatory activity in cultured murine Peyer’s patch cells [28] (Table 1).

4. Gnetin C and Prostate Cancer

Due to its long latency period, prostate cancer is considered an ideal type of cancer for nutritional chemoprevention. Some studies have demonstrated that stilbenes, like other polyphenols, exhibit multitargeted effects in prostate cancer by causing cell cycle arrest/apoptosis and obstructing cell survival and angiogenesis through various mechanisms [31,32]. Our current knowledge about the anticancer activities of stilbenes, including Gnetin C, is largely based on a series of in vitro and preclinical studies in prostate cancer performed by Dr. Anait S. Levenson’s group. These studies were focused on two essential pathways that were regulated by stilbenes: AR-dependent and -independent pathways represented by MTA1-mediated cell survival and metastasis. The results from these studies gave the most compelling evidence for cancer prevention, interception and therapy by stilbenes, showing that both oral and parenteral administration of these compounds was linked to a reduced risk of prostate cancer.
Regarding anti-androgenic mechanisms of action, numerous studies showed that resveratrol and other monomeric stilbenes exert antiproliferative and pro-apoptotic effects in prostate cancer by inhibiting the expression or hindering the function of AR [33,34,35,36,37,38,39,40,41]. Importantly, monomeric stilbenes inhibit not only full-length but also truncated AR, a hallmark of castrate-resistant prostate cancer (CRPC), apparently through different mechanisms compared to traditional non-steroidal antiandrogens, suggesting that combination strategies may result in synergistic effects [37,42,43]. A recently published paper showed that Gnetin C alone and in combination with enzalutamide (Enz) effectively inhibited both full-length and AR splice variant AR-V7 in vitro and in vivo [44] (see more details below).
Metastasis-associated protein is part of the nucleosome remodeling and deacetylation (NuRD) complex, which is involved in gene-specific transcriptional regulation controlling inflammation-driven prostate tumorigenesis and prostate cancer survival pathways and metastasis [45,46]. Earlier experiments with resveratrol demonstrated that it targets MTA1 for degradation in prostate cancer cells and rescues p53 acetylation, resulting in the induction of Bax- and p21-mediated apoptosis [47]. Further, pterostilbene was seven-fold more effective than resveratrol in inhibiting MTA1. It also showed an improved reduction in tumor growth and site metastases in an orthotopic DU145-xenograft mouse model. DU145-MTA1 knockdown-sensitized tumors responded to stilbene treatment with an additional reduction in size, confirming the pleotropic effects of stilbenes [48]. Based on the above findings, subsequent studies with pterostilbene were designed to investigate its effects in prostate-specific Pten-heterozygous (Pten+/f; Cre+) and Pten-null (Ptenf/f; Cre+) transgenic mouse models, which express high prostate levels of MTA1. In the chemoprevention modality (Pten+/f; Cre+), mice were fed a diet supplemented with pterostilbene (100 mg/kg diet), while in the intervention modality (Ptenf/f; Cre+), mice were treated with daily pterostilbene (10 mg/kg bw) i.p. injections. Data showed that pterostilbene treatment inhibited the conversion of high-grade prostate intraepithelial neoplasia (PIN) lesions to cancer by acting through MTA1-mediated signaling to modulate PTEN acetylation, p-Akt/Akt, AR, CyclinD1, NFκB, TGFβ and IL-1β [41]. In combination with histone deacetylase (HDAC) inhibitor suberoylanilide hydroxamic acid (SAHA), pterostilbene showed more potent antitumor effects in Ptenf/f; Cre+ mice, mediated through reduced angiogenesis (MTA1-associated pro-angiogenic factors HIF-1α, VEGF and IL-1β) [49]. Finally, pterostilbene dietary supplementation provided to a prostate-specific MTA1-overexpressing transgenic mouse model mimicking high-risk premalignant prostate cancer (R26MTA1, Pten+/f; Cre+) resulted in an MTA1-mediated (Cyclin D1, Notch2, IL-6, IL-1β, miR-22, miR-34a) reduction in tumor growth and progression, favorable histopathology and reduced angiogenesis, highlighting the potential for oral pterostilbene in prostate cancer [50].
Overall, pterostilbene is recognized as a lead monomeric analog of resveratrol with the most potent anticancer activity in prostate cancer. We observed superior anti-inflammatory (TGFβ, NFκB, IL-1β, IL-2) and anticancer effects of pterostilbene through MTA1 downregulation-associated increase in p53 and PTEN acetylation, inhibition of survival pathways (pAkt/Akt, Notch2, ETS2, CyclinD1, AR), less angiogenesis (VEGF, HIF1α, IL-1β) and a higher apoptotic index (p53, Bax, Bak, cleaved caspase 3).
What about Gnetin C? Gnetin C comparative in vitro experiments with resveratrol and pterostilbene as reference stilbenes showed more potent MTA1 inhibition (in both mRNA and protein levels) and greater MTA1-mediated cytotoxicity in various prostate cancer cells such as DU145, PC3M, 22RV1 and VCAP. Gnetin C displayed substantial inhibitory effects in prostate cancer cells expressing MTA1, while in MTA1 knockdown cells, Gnetin C showed partial MTA1-independent mechanisms that inhibit cell metastatic potential and induce apoptosis. Superior MTA1-mediated anticancer effects of Gnetin C also included a downregulation of oncogenic/tumor-promoting ETS2, pAkt/Akt, Cyclin D1, p-mTOR/pS6K/p4EBP1 and AR signaling [44,51,52,53,54] (Table 2).
Importantly, preclinical animal models, both xenografts and transgenic mice, were used for evaluating Gnetin C’s MTA1-targeted efficacy. First, in vivo effects of Gnetin C in prostate cancer were shown in PC3M-Luc subcutaneous xenografts with i.p. injections [52]. A significant reduction in MTA1-mediated tumor growth and angiogenesis with an induction of apoptosis were observed with Gnetin C (50 mg/kg bw, i.p.). Moreover, the effects of Gnetin C at half the concentration (25 mg/kg bw) were comparable with the antitumor effects of resveratrol and pterostilbene at 50 mg/kg bw, indicating a more potent bioactivity of Gnetin C (Table 2). As confirmation, we found an accumulation of Gnetin C in tumor tissues, while resveratrol and pterostilbene did not reach detectable limits [52]. Further, Gnetin C alone (40 mg/kg bw, i.p.) and combined with Enz (7 mg/kg bw, i.p.) effectively inhibited AR- and MTA1-promoted tumor progression in castrate-resistant 22Rv1-Luc xenografts, resulting in a lower proliferative index (Ki67) and angiogenesis (CD31), and higher apoptosis (CC3) in tumor tissues. Once again, Gnetin C was the most potent inhibitor of AR-V7 and a sensitizer for the Enz response in 22Rv1-resistant cells. It seems that targeting two major pathways in prostate cancer maximizes the efficacy of Gnetin C without compromising its safety profile [44] (Table 2). Finally, two carefully designed preclinical experimental approaches for evaluating Gnetin C’s potential in chemoprevention, interception and therapy in prostate cancer involved transgenic mouse models: one depicting a high-risk premalignant prostate tissues overexpressing MTA1 on the background of Pten heterozygosity (R26MTA1; Pten +/f; Cre+) [54] and another representing MTA1-overexpressing advanced prostate cancer (R26MTA1; Ptenf/f; Cre+) [53]. A Gnetin C-supplemented diet reduced the progression of prostate cancer in high-risk, early-stage prostate tumors by reducing cell proliferation, inflammation and formation of blood vessels. The data also showed a more potent MTA1/PTEN/Akt response to Gnetin C-supplemented diets (35 and 70 mg/kg diet) compared to the reference pterostilbene-supplemented diet (70 mg/kg diet) [54]. Similarly, in an advanced prostate cancer model, Gnetin C (7 mg/kg/bw, i.p.) significantly blocked tumor progression through MTA1/Akt/mTOR signaling [53], suggesting that Gnetin C could be effective not only in cancer chemoprevention and interception but also as an active therapeutic strategy (Table 2).
Studies have shown that stilbenes exhibit cytokine-mediated anti-inflammatory effects in cancer [28,55,56,57,58]. In prostate cancer, the inhibitory effects of pterostilbene on circulating levels of IL-6 and IL-1β were reported in prostate cancer xenografts and transgenic mouse models [49,50]. A low-dose Gnetin C-supplemented diet significantly suppressed levels of pro-inflammatory IL-2 [53,54], and, to a lesser extent, IL-6, in an early-stage prostate cancer model. Interestingly, a high-dose Gnetin C diet had the opposite effect on IL-6 levels [54]. This phenomenon of low-dose natural polyphenols having better efficacy than high doses, when used alone or in combination with other natural agents or chemotherapeutic drugs, has been reported and discussed [17,59,60,61,62]. Further studies are necessary to clarify the dose-and-efficacy outcome of Gnetin C alone and in combination with other agents as anti-inflammatory and anticancer drugs.
To summarize, there are currently only five publications demonstrating the anticancer activity of Gnetin C in different types of cancers, shown mostly in vitro (Table 1). However, comprehensive mechanistic and preclinical studies in prostate cancer support AR- and MTA1-targeted chemopreventive, interceptive and therapeutic effects of Gnetin C.

5. Gnetin C and Other Effects

There are few reports on Gnetin C as a single agent in diseases other than cancer; therefore, studies using MSE, which contains Gnetin C, will also be discussed in this section. Representative studies are outlined in Table 3.
Cardioprotective Effects: Resveratrol was first noted for its cardiovascular benefits from the moderate consumption of red wine, known as “the French paradox”, back in 1992 [69]. Since then, countless studies have confirmed resveratrol’s beneficial effect on atherosclerosis, hypertension, thrombosis, myocardial infarction, heart failure and stroke [70,71,72]. Gnetin C treatment of COS-1 cells resulted in a mild agonistic effect on PPARα and PPARγ, which are involved in HDL-C upregulation. In addition, AT1 receptor binding was significantly inhibited by Gnetin C and MSE [21] (Table 3). Gnetin C was also found to more potently inhibit platelet–collagen adhesion compared to resveratrol [63]. Importantly, two randomized, double-blind, placebo-controlled clinical trials demonstrated the safety and cardioprotective effects of Gnetin C and MSE [21,24] (Table 4).
Neuroprotective Effects: Accumulation of beta-amyloid (Aβ) oligomers is an important factor in neurodegenerative diseases, such as Alzheimer’s disease and other cognitive disorders. Although resveratrol is well-known for its neuroprotective effects, we are only beginning to understand the role that resveratrol dimers can play in neuronal damage. Seino et al. have demonstrated that by reducing Aβ42 secretion, downregulating BACE1 and upregulating MMP-14, Gnetin C may be particularly beneficial in diseases characterized by Aβ accumulation [64] (Table 3). Further, Riviere et al. demonstrated 39% inhibition of Aβ fibril formation by Gnetin C and 63% inhibition by resveratrol [73], while four glucosides of Gnetin C were found to be active inhibitors of Aβ [74]. However, regarding potential neuroprotective effects of stilbenes in Parkinson’s disease and anxiety disorders related to MAO-A- and MAO-B-mediated dopamine and serotonin breakdown, it has been shown that stilbenes act as MAO-A and MAO-B inhibitors. Interestingly, while resveratrol showed preferential MAO-A inhibitory activity [75,76], pterostilbene was the most potent MAO-B inhibitor among 13 tested dietary phenolics, including Gnetin C, which did not show appreciable inhibition of either enzyme [75]. Another study indicated that by negatively regulating IFNβ expression in astrocytoma cells and STAT1 phosphorylation in neuroblastoma cells, Gnetin C could control TLR3-mediated brain inflammation and its neurodegenerative consequences [65]. While there are numerous studies with resveratrol, pterostilbene and other stilbenes in Alzheimer’s disease in vitro and in vivo [77,78,79], to the best of our knowledge, no in vivo studies involving Gnetin C in Alzheimer’s disease or any other neurodegenerative disease are publicly available.
Metabolic Effects: Beneficial effects of MSE in preventing obesity-related disorders and promoting longevity have been shown in mice fed with high-fat diets. Authors found that MSE dose-dependently reduced body weight gain, visceral fat weight and insulin resistance [80].
Non-alcoholic fatty liver diseases (NAFLDs) include a range of conditions such as fatty liver, steatohepatitis, cirrhosis and hepatocellular carcinoma. Advanced cases are often characterized by lipid deposition, inflammation and fibrosis. In their study using a high-fat choline-deficient (HFCD) diet-induced NAFLD mouse model, Kabir et al. found that a diet supplemented with MSE, Gnetin C or resveratrol could significantly reduce body and liver weight, lower plasma triglycerides and non-esterified fatty acids (NEFA) and reduce steatosis and hepatic fibrosis [66] (Table 3). Furthermore, Gnetin C was found to improve blood glucose levels and insulin insensitivity and had a greater lipid-lowering effect compared to resveratrol. In addition, Gnetin C significantly reduced the expression of proteins involved in fatty acid synthesis, transport and lipid metabolism as well as markers in the TGF-β1 signaling pathway (Table 3). Another study revealed the inhibitory effect of MSE on endothelial senescence in streptozotocin-induced (STZ)-diabetic mice. Curiously, despite higher Gnetin C plasma concentrations, in vitro experiments using HUVECs revealed that resveratrol had a greater positive effect on key markers of vascular senescence [16]. Finally, Gnetin C in addition to its antioxidant property showed higher potential to reduce fat absorption and control blood glucose levels compared to resveratrol [15] (Table 3).
Anti-Inflammatory and Anti-Aging Effects: Dermatological age-related changes are often caused by an accumulation of reactive oxygen species (ROS) that promote oxidative damage to cells. The cytoplasmic SOD1 enzyme is an important antioxidant that protects cells by catalyzing superoxide radicals and restoring a healthy redox balance. Watanabe et al. reported that administration of resveratrol- and MSE-supplemented diets in SOD1-deficient mice provide protective effects against skin age-related diseases by upregulating Col1a1 and Sirt1 levels and reducing p53, a known accelerator of skin aging [81]. In addition, these diets reduce other markers of oxidative stress, namely ROS levels in bone marrow and 8-isoprostone in plasma in SOD1-deficient mice [82]. Antioxidant and anti-inflammatory effects of Gnetin C in comparison with resveratrol were demonstrated in a mouse model of periodontitis. Gnetin C induced greater bone healing compared to resveratrol at each time point during eight days of treatment. Furthermore, Gnetin C was superior to resveratrol in inhibiting IL-1β and oxidative stress markers 8-hydroxy-2′-deoxyguanosine and ROS expression. The antioxidative effect of Gnetin C is likely mediated through Nrf2, an important molecule of the oxidative stress response [67,68] (Table 3). Furthermore, Gnetin C was also suggested as a potential safe and effective skin-whitening agent due to its effective inhibition of melanogenesis [27].

6. Gnetin C and Human Clinical Trials

Unfortunately, many clinical trials with natural products (single or combinations) either fail or show conflicting results [59]. Clinical trials with stilbenoids, mainly resveratrol, produced inconsistent data due to differences in trial design, concentrations and formulations used and heterogeneity of the cohort enrolled. The pleotropic effects of these compounds also influence the interpretation of trial results, confounded further by a lack of solid information regarding mechanistic data on pathways and targets (reviewed in [83,84,85]). Considering that a major obstacle of resveratrol is its extensive metabolism and low bioavailability, it would make sense to pursue clinical trials with Gnetin C instead, which shows biologically active concentrations in mice and humans [16,17,22,24]. In addition to its improved bioactivity, Gnetin C is characterized by reduced clearance, a longer mean residence time (MRT) and overall greater systemic exposure compared to resveratrol and pterostilbene [8,22]. This superior pharmacokinetic profile makes Gnetin C a more suitable candidate for clinical trials.
So far, five clinical trials (four with MSE and one with Gnetin C) have been performed in healthy volunteers with the goal of establishing pharmacological safety and the chemopreventive potential [21,22,23,24,25] (Table 4). Randomized, double-blind, placebo-controlled trials were well-tolerated and showed anti-inflammatory, antioxidant and cardioprotective effects of MSE and Gnetin C.
We are only beginning to appreciate the benefits of Gnetin C in humans. Many more well-designed clinical trials are needed to evaluate Gnetin C’s preventive and therapeutic activity in different diseases including obesity, diabetes, cardiovascular disease and cancer. Strategies such as developing more potent Gnetin C derivatives or suitable nanocarriers that may enhance Gnetin C’s activity should be considered.

7. Conclusions

Based on the published scientific data currently available, we believe that there is great promise for Gnetin C as a lead stilbene compound to be considered for therapeutic goals in cancers and for a range of other diseases. Although there is much work yet to be done to reveal the full potential of Gnetin C, substantial information already exists to justify the evaluation of Gnetin C in chemoprevention/interception prostate cancer clinical trials, in which only those patients under active surveillance with MTA1 overexpression are recruited. These patients could benefit considerably from dietary interventions containing Gnetin C. Furthermore, trials where Gnetin C is used in combination with endocrine or chemotherapeutic drugs to sensitize and potentiate anticancer efficacy could be considered for patients with advanced prostate cancer.
With the exception of prostate cancer, there is currently a deficiency in comprehensive preclinical evidence for Gnetin C which delays clinical trials in other diseases and cancer types. In addition, further investigation is needed to explore the pharmacokinetics and pharmacodynamics of Gnetin C used alone or in combination with other agents to maximize chemopreventive and therapeutic efficacy.

Author Contributions

Conceptualization: A.S.L.; Writing—Original Draft Preparation: G.C. and A.S.L.; Writing—Review and Editing: G.C. and A.S.L. Both authors contributed substantially to each section of the text. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We thank Hosoda SHC Co., Ltd. (Fukui, Japan) for providing Gnetin C for our studies.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

Amyloid beta
Aβ42Amyloid beta 42
ACC1Acetyl coenzyme A carboxylase 1
AKTV-akt murine thymoma viral oncogene (protein kinase B)
ALTAlanine aminotransferase
AMLAcute myeloid leukemia
AML-MTAcute myeloid leukemia xenograft
APNAdiponectin
ARAndrogen receptor
AR-FLAndrogen receptor-full length
AR-V7Androgen receptor-variant 7
AsPC1Human pancreatic cancer cells
AT1Angiotensin II type 1 receptor
BACE1Beta-site amyloid precursor protein cleaving enzyme 1
BakHomolog of Bax
BaxBcl-2-associated X protein
Bcl-2B-cell leukemia/lymphoma 2
B16-F10Mouse melanoma cells
BFGF Basic fibroblast growth factor
bwBody weight
CaP8Mouse prostate cancer cells
chREBPCarbohydrate response element binding protein
CC3Cleaved caspase 3
CCL2Chemokine (C-C motif) ligand 2
CCL5Chemokine (C-C motif) ligand 5
CD31 Cluster of differentiation 31
CMLChronic myeloid leukemia
Col1a1Collagen type I alpha 1 chain
COS-1Monkey kidney cells
CreCre recombinase
CRPC-reactive protein
CRPCCastrate-resistant prostate cancer
CTLCytotoxic T lymphocytes
DGAT1 Diacylglycerol acyltransferase 1
DGAT2Diacylglycerol acyltransferase 2
DU145Human prostate cancer cells
eNOSEndothelial nitric oxide synthase
EnzEnzalutamide
ERK1/2Mitogen-activated protein kinase 1 and 2
ETS2ETS Proto-oncogene 2, transcription factor
GnCGnetin C
HDACHistone deacetylase
HDL-CHigh-density lipoprotein cholesterol
HEK-293THuman kidney cells
HFCDHigh-fat choline deficient
HIF-1αHypoxia-inducible factor 1- alpha
HL60Human leukemia cells
HT-29Human colon cancer cells
HUVECHuman umbilical vein endothelial cells
IFN-βInterferon beta
IFN-γInterferon gamma
IL-1βInterleukin-1 beta
IL-2Interleukin 2
IL-6Interleukin 6
i.pIntraperitoneal
K562Human CML cells
KH88Human CML cells
Ki67Cellular protein marker of proliferation
LDHLactate dehydrogenase
LDL-CLow-density lipoprotein-cholesterol
LDLRLow-density lipoprotein receptor
LNCaPHuman prostate cancer cells
LucLuciferase
MAO-AMonoamine oxidase A
MAO-BMonoamine oxidase B
MCF7Human breast cancer cells
miR-22MicroRNA-22
miR-34aMicroRNA-34a
MMP-14Matrix metallopeptidase 14
MRTMean residence time
MSEMelinjo seed extract
MTA1Metastasis-associated protein 1
MTPMicrosomal triglyceride transfer protein
mTORMammalian target of rapamycin
MV4Human AML cells
NAFLDNonalcoholic fatty liver disease
NEFANonesterified fatty acids
NF- κβNuclear factor kappa-light-chain-enhancer of activated B cells
NKNatural killer cells
Nrf2Nuclear factor erythroid 2-related factor 2
NuRDNucleosome remodeling deacetylation complex
Oun1Human CML cells
p21Cyclin-dependent kinase inhibitor 1A
p4EBP1Eukaryotic translation initiation factor 4E-binding protein 1
p53Tumor protein p53
PAI1Plasminogen activator inhibitor-1
p-AKTPhosphorylated AKT
Pan-02Mouse pancreatic cancer cells
PANC-1Human pancreatic cancer cells
PC3Human prostate cancer cells
PC3MHuman prostate cancer cells
PGC-1aperoxisome proliferator-activated receptor gamma coactivator 1 alpha
PINProstatic intraepithelial neoplasia
PPARαPeroxisome proliferator-activated receptor alpha
PPARγPeroxisome proliferator-activated receptor gamma
pS6Kp70-S6 Kinase
PSAProstate specific antigen
PTENPhosphatase and tensin homolog
PWPE1normal prostate epithelial cells
R26Rosa26
ResResveratrol
ROSReactive oxygen species
22Rv1Human prostate cancer cells
SA-β-galSenescence-associated beta-galactosidase
SAHASuberoylanilide hydroxamic acid
shMTA1Short-hairpin MTA1
Sirt1Sirtuin 1
SOD1Superoxide dismutase 1
STAT1Signal transducer and activator of transcription 1
STZStreptozotocin
TgTriglyceride
TGFβTransforming growth factor beta
THP1Human leukemia cells
TLR3Toll-like receptor 3
U937Human lymphoma cells
VCAM-1Vascular cell adhesion molecule-1
VCaPHuman prostate cancer cells
VEGFVascular epidermal growth factor
WTWild type

References

  1. Ahmadi, R.; Ebrahimzadeh, M.A. Resveratrol—A comprehensive review of recent advances in anticancer drug design and development. Eur. J. Med. Chem. 2020, 200, 112356. [Google Scholar] [CrossRef] [PubMed]
  2. Li, Y.R.; Li, S.; Lin, C.C. Effect of resveratrol and pterostilbene on aging and longevity. Biofactors 2018, 44, 69–82. [Google Scholar] [CrossRef] [PubMed]
  3. Lange, K.W.; Li, S. Resveratrol, pterostilbene, and dementia. Biofactors 2018, 44, 83–90. [Google Scholar] [CrossRef] [PubMed]
  4. Kim, H.; Seo, K.H.; Yokoyama, W. Chemistry of Pterostilbene and Its Metabolic Effects. J. Agric. Food Chem. 2020, 68, 12836–12841. [Google Scholar] [CrossRef]
  5. Asensi, M.; Ortega, A.; Mena, S.; Feddi, F.; Estrela, J.M. Natural polyphenols in cancer therapy. Crit. Rev. Clin. Lab. Sci. 2011, 48, 197–216. [Google Scholar] [CrossRef]
  6. Miksits, M.; Wlcek, K.; Svoboda, M.; Kunert, O.; Haslinger, E.; Thalhammer, T.; Szekeres, T.; Jager, W. Antitumor activity of resveratrol and its sulfated metabolites against human breast cancer cells. Planta Med. 2009, 75, 1227–1230. [Google Scholar] [CrossRef]
  7. Hoshino, J.; Park, E.J.; Kondratyuk, T.P.; Marler, L.; Pezzuto, J.M.; van Breemen, R.B.; Mo, S.; Li, Y.; Cushman, M. Selective synthesis and biological evaluation of sulfate-conjugated resveratrol metabolites. J. Med. Chem. 2010, 53, 5033–5043. [Google Scholar] [CrossRef]
  8. Kapetanovic, I.M.; Muzzio, M.; Huang, Z.; Thompson, T.N.; McCormick, D.L. Pharmacokinetics, oral bioavailability, and metabolic profile of resveratrol and its dimethylether analog, pterostilbene, in rats. Cancer Chemother. Pharmacol. 2011, 68, 593–601. [Google Scholar] [CrossRef]
  9. Ito, T. Resveratrol oligomer structure in Dipterocarpaceaeous plants. J. Nat. Med. 2020, 74, 619–637. [Google Scholar] [CrossRef]
  10. Shen, J.; Zhou, Q.; Li, P.; Wang, Z.; Liu, S.; He, C.; Zhang, C.; Xiao, P. Update on Phytochemistry and Pharmacology of Naturally Occurring Resveratrol Oligomers. Molecules 2017, 22, 2050. [Google Scholar] [CrossRef]
  11. Kang, J.E.; Yoo, N.; Jeon, B.J.; Kim, B.S.; Chung, E.H. Resveratrol Oligomers, Plant-Produced Natural Products With Anti-virulence and Plant Immune-Priming Roles. Front. Plant Sci. 2022, 13, 885625. [Google Scholar] [CrossRef] [PubMed]
  12. Liu, S.; Li, Y.; Yi, F.; Liu, Q.; Chen, N.; He, X.; He, C.; Xiao, P. Resveratrol oligomers from Paeonia suffruticosa protect mice against cognitive dysfunction by regulating cholinergic, antioxidant and anti-inflammatory pathways. J. Ethnopharmacol. 2020, 260, 112983. [Google Scholar] [CrossRef]
  13. Gonzalez-Sarrias, A.; Gromek, S.; Niesen, D.; Seeram, N.P.; Henry, G.E. Resveratrol oligomers isolated from Carex species inhibit growth of human colon tumorigenic cells mediated by cell cycle arrest. J. Agric. Food Chem. 2011, 59, 8632–8638. [Google Scholar] [CrossRef] [PubMed]
  14. Xue, Y.Q.; Di, J.M.; Luo, Y.; Cheng, K.J.; Wei, X.; Shi, Z. Resveratrol oligomers for the prevention and treatment of cancers. Oxid. Med. Cell. Longev. 2014, 2014, 765832. [Google Scholar] [CrossRef] [PubMed]
  15. Kato, E.; Tokunaga, Y.; Sakan, F. Stilbenoids isolated from the seeds of Melinjo (Gnetum gnemon L.) and their biological activity. J. Agric. Food Chem. 2009, 57, 2544–2549. [Google Scholar] [CrossRef]
  16. Ota, H.; Akishita, M.; Tani, H.; Tatefuji, T.; Ogawa, S.; Iijima, K.; Eto, M.; Shirasawa, T.; Ouchi, Y. trans-Resveratrol in Gnetum gnemon protects against oxidative-stress-induced endothelial senescence. J. Nat. Prod. 2013, 76, 1242–1247. [Google Scholar] [CrossRef]
  17. Narayanan, N.K.; Kunimasa, K.; Yamori, Y.; Mori, M.; Mori, H.; Nakamura, K.; Miller, G.; Manne, U.; Tiwari, A.K.; Narayanan, B. Antitumor activity of melinjo (Gnetum gnemon L.) seed extract in human and murine tumor models in vitro and in a colon-26 tumor-bearing mouse model in vivo. Cancer Med. 2015, 4, 1767–1780. [Google Scholar] [CrossRef]
  18. Espinoza, J.L.; Elbadry, M.I.; Taniwaki, M.; Harada, K.; Trung, L.Q.; Nakagawa, N.; Takami, A.; Ishiyama, K.; Yamauchi, T.; Takenaka, K.; et al. The simultaneous inhibition of the mTOR and MAPK pathways with Gnetin-C induces apoptosis in acute myeloid leukemia. Cancer Lett. 2017, 400, 127–136. [Google Scholar] [CrossRef]
  19. Kunimasa, K.; Ohta, T.; Tani, H.; Kato, E.; Eguchi, R.; Kaji, K.; Ikeda, K.; Mori, H.; Mori, M.; Tatefuji, T.; et al. Resveratrol derivative-rich melinjo (Gnetum gnemon L.) seed extract suppresses multiple angiogenesis-related endothelial cell functions and tumor angiogenesis. Mol. Nutr. Food Res. 2011, 55, 1730–1734. [Google Scholar] [CrossRef]
  20. Tatefuji, T.; Yanagihara, M.; Fukushima, S.; Hashimoto, K. Safety assessment of melinjo (Gnetum gnemon L.) seed extract: Acute and subchronic toxicity studies. Food Chem. Toxicol. 2014, 67, 230–235. [Google Scholar] [CrossRef]
  21. Konno, H.; Kanai, Y.; Katagiri, M.; Watanabe, T.; Mori, A.; Ikuta, T.; Tani, H.; Fukushima, S.; Tatefuji, T.; Shirasawa, T. Melinjo (Gnetum gnemon L.) Seed Extract Decreases Serum Uric Acid Levels in Nonobese Japanese Males: A Randomized Controlled Study. Evid. Based Complement. Alternat Med. 2013, 2013, 589169. [Google Scholar] [CrossRef] [PubMed]
  22. Tani, H.; Hikami, S.; Iizuna, S.; Yoshimatsu, M.; Asama, T.; Ota, H.; Kimura, Y.; Tatefuji, T.; Hashimoto, K.; Higaki, K. Pharmacokinetics and safety of resveratrol derivatives in humans after oral administration of melinjo (Gnetum gnemon L.) seed extract powder. J. Agric. Food Chem. 2014, 62, 1999–2007. [Google Scholar] [CrossRef]
  23. Espinoza, J.L.; An, D.T.; Trung, L.Q.; Yamada, K.; Nakao, S.; Takami, A. Stilbene derivatives from melinjo extract have antioxidant and immune modulatory effects in healthy individuals. Integr. Mol. Med. 2015, 2, 405–413. [Google Scholar]
  24. Nakagami, Y.; Suzuki, S.; Espinoza, J.L.; Vu Quang, L.; Enomoto, M.; Takasugi, S.; Nakamura, A.; Nakayama, T.; Tani, H.; Hanamura, I.; et al. Immunomodulatory and Metabolic Changes after Gnetin-C Supplementation in Humans. Nutrients 2019, 11, 1403. [Google Scholar] [CrossRef]
  25. Oniki, K.; Kawakami, T.; Nakashima, A.; Miyata, K.; Watanabe, T.; Fujikawa, H.; Nakashima, R.; Nasu, A.; Eto, Y.; Takahashi, N.; et al. Melinjo seed extract increases adiponectin multimerization in physiological and pathological conditions. Sci. Rep. 2020, 10, 4313. [Google Scholar] [CrossRef] [PubMed]
  26. Espinoza, J.L.; Inaoka, P.T. Gnetin-C and other resveratrol oligomers with cancer chemopreventive potential. Ann. N. Y. Acad. Sci. 2017, 1403, 5–14. [Google Scholar] [CrossRef]
  27. Yanagihara, M.; Yoshimatsu, M.; Inoue, A.; Kanno, T.; Tatefuji, T.; Hashimoto, K. Inhibitory effect of gnetin C, a resveratrol dimer from melinjo (Gnetum gnemon), on tyrosinase activity and melanin biosynthesis. Biol. Pharm. Bull. 2012, 35, 993–996. [Google Scholar] [CrossRef]
  28. Kato, H.; Samizo, M.; Kawabata, R.; Takano, F.; Ohta, T. Stilbenoids from the melinjo (Gnetum gnemon L.) fruit modulate cytokine production in murine Peyer’s patch cells ex vivo. Planta Med. 2011, 77, 1027–1034. [Google Scholar] [CrossRef]
  29. Iliya, I.; Akao, Y.; Matsumoto, K.; Nakagawa, Y.; Zulfiqar, A.; Ito, T.; Oyama, M.; Murata, H.; Tanaka, T.; Nozawa, Y.; et al. Growth inhibition of stilbenoids in Welwitschiaceae and Gnetaceae through induction of apoptosis in human leukemia HL60 cells. Biol. Pharm. Bull. 2006, 29, 1490–1492. [Google Scholar] [CrossRef]
  30. Ferrer, P.; Asensi, M.; Segarra, R.; Ortega, A.; Benlloch, M.; Obrador, E.; Varea, M.T.; Asensio, G.; Jorda, L.; Estrela, J.M. Association between pterostilbene and quercetin inhibits metastatic activity of B16 melanoma. Neoplasia 2005, 7, 37–47. [Google Scholar] [CrossRef]
  31. Oczkowski, M.; Dziendzikowska, K.; Pasternak-Winiarska, A.; Wlodarek, D.; Gromadzka-Ostrowska, J. Dietary Factors and Prostate Cancer Development, Progression, and Reduction. Nutrients 2021, 13, 496. [Google Scholar] [CrossRef] [PubMed]
  32. Lin, V.C.; Tsai, Y.C.; Lin, J.N.; Fan, L.L.; Pan, M.H.; Ho, C.T.; Wu, J.Y.; Way, T.D. Activation of AMPK by pterostilbene suppresses lipogenesis and cell-cycle progression in p53 positive and negative human prostate cancer cells. J. Agric. Food Chem. 2012, 60, 6399–6407. [Google Scholar] [CrossRef] [PubMed]
  33. Harada, N.; Atarashi, K.; Murata, Y.; Yamaji, R.; Nakano, Y.; Inui, H. Inhibitory mechanisms of the transcriptional activity of androgen receptor by resveratrol: Implication of DNA binding and acetylation of the receptor. J. Steroid Biochem. Mol. Biol. 2011, 123, 65–70. [Google Scholar] [CrossRef]
  34. Mitchell, S.H.; Zhu, W.; Young, C.Y. Resveratrol inhibits the expression and function of the androgen receptor in LNCaP prostate cancer cells. Cancer Res. 1999, 59, 5892–5895. [Google Scholar] [PubMed]
  35. Harada, N.; Murata, Y.; Yamaji, R.; Miura, T.; Inui, H.; Nakano, Y. Resveratrol down-regulates the androgen receptor at the post-translational level in prostate cancer cells. J. Nutr. Sci. Vitaminol. 2007, 53, 556–560. [Google Scholar] [CrossRef]
  36. Wang, Y.; Romigh, T.; He, X.; Orloff, M.S.; Silverman, R.H.; Heston, W.D.; Eng, C. Resveratrol regulates the PTEN/AKT pathway through androgen receptor-dependent and -independent mechanisms in prostate cancer cell lines. Hum. Mol. Genet. 2010, 19, 4319–4329. [Google Scholar] [CrossRef]
  37. Streicher, W.; Luedeke, M.; Azoitei, A.; Zengerling, F.; Herweg, A.; Genze, F.; Schrader, M.G.; Schrader, A.J.; Cronauer, M.V. Stilbene induced inhibition of androgen receptor dimerization: Implications for AR and ARDeltaLBD-signalling in human prostate cancer cells. PLoS ONE 2014, 9, e98566. [Google Scholar] [CrossRef]
  38. Chakraborty, S.; Kumar, A.; Butt, N.A.; Zhang, L.; Williams, R.; Rimando, A.M.; Biswas, P.K.; Levenson, A.S. Molecular insight into the differential anti-androgenic activity of resveratrol and its natural analogs: In silico approach to understand biological actions. Mol. Biosyst. 2016, 12, 1702–1709. [Google Scholar] [CrossRef]
  39. Nikhil, K.; Sharan, S.; Chakraborty, A.; Roy, P. Pterostilbene-isothiocyanate conjugate suppresses growth of prostate cancer cells irrespective of androgen receptor status. PLoS ONE 2014, 9, e93335. [Google Scholar] [CrossRef]
  40. Lundqvist, J.; Tringali, C.; Oskarsson, A. Resveratrol, piceatannol and analogs inhibit activation of both wild-type and T877A mutant androgen receptor. J. Steroid Biochem. Mol. Biol. 2017, 174, 161–168. [Google Scholar] [CrossRef]
  41. Dhar, S.; Kumar, A.; Zhang, L.; Rimando, A.M.; Lage, J.M.; Lewin, J.R.; Atfi, A.; Zhang, X.; Levenson, A.S. Dietary pterostilbene is a novel MTA1-targeted chemopreventive and therapeutic agent in prostate cancer. Oncotarget 2016, 7, 18469–18484. [Google Scholar] [CrossRef]
  42. Kai, L.; Levenson, A.S. Combination of resveratrol and antiandrogen flutamide has synergistic effect on androgen receptor inhibition in prostate cancer cells. Anticancer. Res. 2011, 31, 3323–3330. [Google Scholar] [PubMed]
  43. Kumar, A.; Lin, S.-Y.; Dhar, S.; Rimando, A.M.; Levenson, A.S. Stilbenes inhibit androgen receptor expression in 22Rv1 castrate-resistant prostate cancer cells. J. Med. Act. Plants 2014, 3, 1–8. [Google Scholar]
  44. Campanelli, G.; Deabel, R.A.; Puaar, A.; Devarakonda, L.S.; Parupathi, P.; Zhang, J.; Waxner, N.; Yang, C.; Kumar, A.; Levenson, A.S. Molecular Efficacy of Gnetin C as Dual-Targeted Therapy for Castrate-Resistant Prostate Cancer. Mol. Nutr. Food Res. 2023, 67, e2300479. [Google Scholar] [CrossRef]
  45. Ma, J.; Li, C.; Qian, H.; Zhang, Y. MTA1: A Vital Modulator in Prostate Cancer. Curr. Protein Pept. Sci. 2022, 23, 456–464. [Google Scholar]
  46. Sen, N.; Gui, B.; Kumar, R. Role of MTA1 in cancer progression and metastasis. Cancer Metastasis Rev. 2014, 33, 879–889. [Google Scholar] [CrossRef] [PubMed]
  47. Kai, L.; Samuel, S.K.; Levenson, A.S. Resveratrol enhances p53 acetylation and apoptosis in prostate cancer by inhibiting MTA1/NuRD complex. Int. J. Cancer 2010, 126, 1538–1548. [Google Scholar] [CrossRef]
  48. Li, K.; Dias, S.J.; Rimando, A.M.; Dhar, S.; Mizuno, C.S.; Penman, A.D.; Lewin, J.R.; Levenson, A.S. Pterostilbene acts through metastasis-associated protein 1 to inhibit tumor growth, progression and metastasis in prostate cancer. PLoS ONE 2013, 8, e57542. [Google Scholar] [CrossRef]
  49. Butt, N.A.; Kumar, A.; Dhar, S.; Rimando, A.M.; Akhtar, I.; Hancock, J.C.; Lage, J.M.; Pound, C.R.; Lewin, J.R.; Gomez, C.R.; et al. Targeting MTA1/HIF-1alpha signaling by pterostilbene in combination with histone deacetylase inhibitor attenuates prostate cancer progression. Cancer Med. 2017, 6, 2673–2685. [Google Scholar] [CrossRef]
  50. Hemani, R.; Patel, I.; Inamdar, N.; Campanelli, G.; Donovan, V.; Kumar, A.; Levenson, A.S. Dietary Pterostilbene for MTA1-Targeted Interception in High-Risk Premalignant Prostate Cancer. Cancer Prev. Res. 2022, 15, 87–100. [Google Scholar] [CrossRef]
  51. Kumar, A.; Dholakia, K.; Sikorska, G.; Martinez, L.A.; Levenson, A.S. MTA1-Dependent Anticancer Activity of Gnetin C in Prostate Cancer. Nutrients 2019, 11, 2096. [Google Scholar] [CrossRef]
  52. Gadkari, K.; Kolhatkar, U.; Hemani, R.; Campanelli, G.; Cai, Q.; Kumar, A.; Levenson, A.S. Therapeutic Potential of Gnetin C in Prostate Cancer: A Pre-Clinical Study. Nutrients 2020, 12, 3631–3642. [Google Scholar] [CrossRef]
  53. Campanelli, G.; Francois, E.; Parupathi, P.; Devarakonda, L.S.; Yang, C.; Kumar, A.; Levenson, A.S. The Therapeutic Efficacy and Mechanism of Action of Gnetin C, a Natural Compound from the Melinjo Plant, in a Preclinical Mouse Model of Advanced Prostate Cancer. Cancers 2024, 16, 1344. [Google Scholar] [CrossRef]
  54. Parupathi, P.; Campanelli, G.; Deabel, R.A.; Puaar, A.; Devarakonda, L.S.; Kumar, A.; Levenson, A.S. Gnetin C Intercepts MTA1-Associated Neoplastic Progression in Prostate Cancer. Cancers 2022, 14, 6038. [Google Scholar] [CrossRef] [PubMed]
  55. Lee, J.; Jung, E.; Lim, J.; Lee, J.; Hur, S.; Kim, S.S.; Lim, S.; Hyun, C.G.; Kim, Y.S.; Park, D. Involvement of nuclear factor-kappaB in the inhibition of pro-inflammatory mediators by pinosylvin. Planta Med. 2006, 72, 801–806. [Google Scholar] [CrossRef]
  56. Clarke, J.O.; Mullin, G.E. A review of complementary and alternative approaches to immunomodulation. Nutr. Clin. Pract. 2008, 23, 49–62. [Google Scholar] [CrossRef]
  57. Pradhan, R.; Chatterjee, S.; Hembram, K.C.; Sethy, C.; Mandal, M.; Kundu, C.N. Nano formulated Resveratrol inhibits metastasis and angiogenesis by reducing inflammatory cytokines in oral cancer cells by targeting tumor associated macrophages. J. Nutr. Biochem. 2021, 92, 108624. [Google Scholar] [CrossRef]
  58. Vo, N.T.; Madlener, S.; Bago-Horvath, Z.; Herbacek, I.; Stark, N.; Gridling, M.; Probst, P.; Giessrigl, B.; Bauer, S.; Vonach, C.; et al. Pro- and anticarcinogenic mechanisms of piceatannol are activated dose dependently in MCF-7 breast cancer cells. Carcinogenesis 2010, 31, 2074–2081. [Google Scholar] [CrossRef]
  59. Sauter, E.R.; Mohammed, A. Natural Products for Cancer Prevention and Interception: Preclinical and Clinical Studies and Funding Opportunities. Pharmaceuticals 2024, 17, 136. [Google Scholar] [CrossRef]
  60. Narisawa, T.; Fukaura, Y.; Hasebe, M.; Nomura, S.; Oshima, S.; Inakuma, T. Prevention of N-methylnitrosourea-induced colon carcinogenesis in rats by oxygenated carotenoid capsanthin and capsanthin-rich paprika juice. Proc. Soc. Exp. Biol. Med. 2000, 224, 116–122. [Google Scholar]
  61. Kresty, L.A.; Morse, M.A.; Morgan, C.; Carlton, P.S.; Lu, J.; Gupta, A.; Blackwood, M.; Stoner, G.D. Chemoprevention of esophageal tumorigenesis by dietary administration of lyophilized black raspberries. Cancer Res. 2001, 61, 6112–6119. [Google Scholar]
  62. Bouayed, J.; Bohn, T. Exogenous antioxidants--Double-edged swords in cellular redox state: Health beneficial effects at physiologic doses versus deleterious effects at high doses. Oxid. Med. Cell. Longev. 2010, 3, 228–237. [Google Scholar] [CrossRef]
  63. Kloypan, C.; Jeenapongsa, R.; Sri-in, P.; Chanta, S.; Dokpuang, D.; Tip-pyang, S.; Surapinit, N. Stilbenoids from Gnetum macrostachyum attenuate human platelet aggregation and adhesion. Phytother. Res. 2012, 26, 1564–1568. [Google Scholar] [CrossRef]
  64. Seino, S.; Kimoto, T.; Yoshida, H.; Tanji, K.; Matsumiya, T.; Hayakari, R.; Seya, K.; Kawaguchi, S.; Tsuruga, K.; Tanaka, H.; et al. Gnetin C, a resveratrol dimer, reduces amyloid-beta 1-42 (Abeta42) production and ameliorates Abeta42-lowered cell viability in cultured SH-SY5Y human neuroblastoma cells. Biomed. Res. 2018, 39, 105–115. [Google Scholar] [CrossRef]
  65. Yoshida, H.; Imaizumi, T.; Matsumiya, T.; Seya, K.; Kawaguchi, S.; Tanaka, H. Gnetin C suppresses double-stranded RNA-induced C-C motif chemokine ligand 2 (CCL2) and CCL5 production by inhibiting Toll-like receptor 3 signaling pathway. Biomed. Res. 2018, 39, 231–240. [Google Scholar] [CrossRef]
  66. Kabir, T.; Yoshiba, H.; Agista, A.Z.; Sultana, H.; Ohsaki, Y.; Yeh, C.L.; Hirakawa, R.; Tani, H.; Ikuta, T.; Nochi, T.; et al. Protective Effects of Gnetin C from Melinjo Seed Extract against High-Fat Diet-Induced Hepatic Steatosis and Liver Fibrosis in NAFLD Mice Model. Nutrients 2023, 15, 3888. [Google Scholar] [CrossRef]
  67. Ikeda, E.; Tanaka, D.; Glogauer, M.; Tenenbaum, H.C.; Ikeda, Y. Healing effects of monomer and dimer resveratrol in a mouse periodontitis model. BMC Oral Health 2022, 22, 460. [Google Scholar] [CrossRef]
  68. Ikeda, E.; Ikeda, Y.; Wang, Y.; Fine, N.; Sheikh, Z.; Viniegra, A.; Barzilay, O.; Ganss, B.; Tenenbaum, H.C.; Glogauer, M. Resveratrol derivative-rich melinjo seed extract induces healing in a murine model of established periodontitis. J. Periodontol. 2018, 89, 586–595. [Google Scholar] [CrossRef]
  69. Renaud, S.; de Lorgeril, M. Wine, alcohol, platelets, and the French paradox for coronary heart disease. Lancet 1992, 339, 1523–1526. [Google Scholar] [CrossRef] [PubMed]
  70. Lippi, G.; Franchini, M.; Favaloro, E.J.; Targher, G. Moderate red wine consumption and cardiovascular disease risk: Beyond the “French paradox”. Semin. Thromb. Hemost. 2010, 36, 59–70. [Google Scholar] [CrossRef] [PubMed]
  71. Bonnefont-Rousselot, D. Resveratrol and Cardiovascular Diseases. Nutrients 2016, 8, 250. [Google Scholar] [CrossRef]
  72. Guo, S.; Zhou, Y.; Xie, X. Resveratrol inhibiting TGF/ERK signaling pathway can improve atherosclerosis: Backgrounds, mechanisms and effects. Biomed. Pharmacother. 2022, 155, 113775. [Google Scholar] [CrossRef]
  73. Riviere, C.; Papastamoulis, Y.; Fortin, P.Y.; Delchier, N.; Andriamanarivo, S.; Waffo-Teguo, P.; Kapche, G.D.; Amira-Guebalia, H.; Delaunay, J.C.; Merillon, J.M.; et al. New stilbene dimers against amyloid fibril formation. Bioorg Med. Chem. Lett. 2010, 20, 3441–3443. [Google Scholar] [CrossRef]
  74. Buffeteau, T.; Cavagnat, D.; Bisson, J.; Marchal, A.; Kapche, G.D.; Battistini, I.; Da Costa, G.; Badoc, A.; Monti, J.P.; Merillon, J.M.; et al. Unambiguous Determination of the Absolute Configuration of Dimeric Stilbene Glucosides from the Rhizomes of Gnetum africanum. J. Nat. Prod. 2014, 77, 1981–1985. [Google Scholar] [CrossRef]
  75. Zhang, Z.; Hamada, H.; Gerk, P.M. Selectivity of Dietary Phenolics for Inhibition of Human Monoamine Oxidases A and B. Biomed. Res. Int. 2019, 2019, 8361858. [Google Scholar] [CrossRef]
  76. Herraiz, T.; Flores, A.; Fernandez, L. Analysis of monoamine oxidase (MAO) enzymatic activity by high-performance liquid chromatography-diode array detection combined with an assay of oxidation with a peroxidase and its application to MAO inhibitors from foods and plants. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2018, 1073, 136–144. [Google Scholar] [CrossRef]
  77. Freyssin, A.; Page, G.; Fauconneau, B.; Rioux Bilan, A. Natural stilbenes effects in animal models of Alzheimer’s disease. Neural Regen. Res. 2020, 15, 843–849. [Google Scholar]
  78. Wicinski, M.; Domanowska, A.; Wodkiewicz, E.; Malinowski, B. Neuroprotective Properties of Resveratrol and Its Derivatives-Influence on Potential Mechanisms Leading to the Development of Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 2749. [Google Scholar] [CrossRef]
  79. Richard, T.; Pawlus, A.D.; Iglesias, M.L.; Pedrot, E.; Waffo-Teguo, P.; Merillon, J.M.; Monti, J.P. Neuroprotective properties of resveratrol and derivatives. Ann. N. Y. Acad. Sci. 2011, 1215, 103–108. [Google Scholar] [CrossRef] [PubMed]
  80. Ikuta, T.; Saito, S.; Tani, H.; Tatefuji, T.; Hashimoto, K. Resveratrol derivative-rich melinjo (Gnetum gnemon L.) seed extract improves obesity and survival of C57BL/6 mice fed a high-fat diet. Biosci. Biotechnol. Biochem. 2015, 79, 2044–2049. [Google Scholar] [CrossRef]
  81. Rodier, F.; Campisi, J.; Bhaumik, D. Two faces of p53: Aging and tumor suppression. Nucleic Acids Res. 2007, 35, 7475–7484. [Google Scholar] [CrossRef] [PubMed]
  82. Watanabe, K.; Shibuya, S.; Ozawa, Y.; Izuo, N.; Shimizu, T. Resveratrol Derivative-Rich Melinjo Seed Extract Attenuates Skin Atrophy in Sod1-Deficient Mice. Oxid. Med. Cell. Longev. 2015, 2015, 391075. [Google Scholar] [CrossRef] [PubMed]
  83. Tome-Carneiro, J.; Larrosa, M.; Gonzalez-Sarrias, A.; Tomas-Barberan, F.A.; Garcia-Conesa, M.T.; Espin, J.C. Resveratrol and clinical trials: The crossroad from in vitro studies to human evidence. Curr. Pharm. Des. 2013, 19, 6064–6093. [Google Scholar] [CrossRef] [PubMed]
  84. Poulsen, M.M.; Fjeldborg, K.; Ornstrup, M.J.; Kjaer, T.N.; Nohr, M.K.; Pedersen, S.B. Resveratrol and inflammation: Challenges in translating pre-clinical findings to improved patient outcomes. Biochim. Biophys. Acta 2015, 1852, 1124–1136. [Google Scholar] [CrossRef]
  85. Dvorakova, M.; Landa, P. Anti-inflammatory activity of natural stilbenoids: A review. Pharmacol. Res. 2017, 124, 126–145. [Google Scholar] [CrossRef]
Figure 1. Image of melinjo plant (Gnetum gnemon). Chemical structures of Gnetin C and stilbene monomers: resveratrol and pterostilbene.
Figure 1. Image of melinjo plant (Gnetum gnemon). Chemical structures of Gnetin C and stilbene monomers: resveratrol and pterostilbene.
Nutrients 17 00863 g001
Table 1. Anticancer activity of Gnetin C.
Table 1. Anticancer activity of Gnetin C.
CompoundModelDoseCell/AnimalMechanism of ActionRef
Gnetin CIn vitro0–100 µMAcute myelogenous leukemia (AML) cells: MV4, THP1, U937, HL60
Chronic myeloid leukemia (CML) cells: K562, Oun1, KH88
Inhibition of ERK 1/2 and AKT/mTOR
pathways;
cell cycle arrest
[18]
In vivo5 mg/kg/day,
5 weeks
AML-MT xenograft miceInhibition of leukemia; antitumor effects (blood, spleen, bone marrow); extended survival of mice
Gnetin C
MSE
Resveratrol
Gnemoside A, C, D
In vitro0.5–10 µM
40 µg/mL
5 µM
Human umbilical vein endothelial cells (HUVECs)Inhibition of tube formation stimulated with VEGF and BFGF; reduction of cell viability and migration; ERK1/2 inactivation[19]
In vivo5% MSEMouse dorsal air sac assayInhibition of tumor angiogenesis
Gnetin C
Resveratrol
MSE
In vitro0–100 µM
0–400 µg/mL
LNCaP, PC3, Murine CaP8 prostate cancer cells;
MCF7 breast cancer cells;
HT-29, colon-26 colon cancer cells;
PANC-1, AsPC1, Pan-02 pancreatic cancer cells;
PWPE1 and HEK-293T cells
Inhibitory effects on cancer cells without affecting normal cells; induction of apoptosis via caspase 3/7-dependent mechanisms[17]
MSEIn vivo50 and 100 mg/kg/day, oralColon-26 tumor-bearing mouse modelInhibition of tumor growth, angiogenesis and liver metastasis
Gnetin C
Resveratrol
In vitro2–16 µMMurine melanoma B16 cellsInhibitory activity against tyrosinase and melanin biosynthesis[27]
Gnetin C
Melinjo fruit extract
Ex vivo50% extract
at 100 mg/kg/day
Cultured murine Peyer’s patch cellsEnhanced T-cell-dependent immune responses, IL2↑, IFNγ↑[28]
↑ upregulation; MSE, melinjo seed extract. LNCaP (AR-positive); PC3 (AR-negative); MCF7 (ER-positive).
Table 2. Gnetin C and prostate cancer.
Table 2. Gnetin C and prostate cancer.
CompoundModelDoseMechanism of ActionRef
MechanisticIn vitroGnetin C
Resveratrol
DU145, PC3M, PC3M-shMTA1, DU145-shMTA10–50 µMMTA1-mediated inhibitory effects on cell viability, colony formation, migration, induction of apoptosis;
MTA1↓ (protein and RNA), ETS2↓
[51]
Gnetin C
Resveratrol
Pterostilbene
DU145, PC3M0–100 µMCytotoxicity, reduction of clonogenic survival and motility[52]
Gnetin C
Enzalutamide
combination
22Rv1, VCaP0–100 µM/
0–50 µM
Inhibition of cell viability, clonogenic survival and migration, synergism at certain doses;
MTA1↓, AR-FL↓, AR-V7↓, PSA↓
[44]
Gnetin C
Pterostilbene
22Rv125 µMMTA1↓, pAkt/Akt↓, PTEN↑[54]
Gnetin CPC3M,
PC3M-shMTA1
25, 50 µMMTA1↓, Cyclin D1↓, pAkt/Akt↓,
p-mTOR/pS6K/p4EBP1↓
[53]
PreclinicalXenograftsGnetin C
Resveratrol
Pterostilbene
PC3M-Luc25 and 50 mg/kg/day, i.p.Tumor growth reduction, inhibition of angiogenesis and induction of apoptosis;
MTA1↓, CyclinD1↓, Notch2↓
[52]
Gnetin C
Enzalutamide
combination
22Rv1-LucGnC 40 mg/kg/day + Enz
7 or 10 mg/kg/day, i.p.
Inhibition of tumor growth and angiogenesis, induction of apoptosis; MTA1↓, AR-FL↓, AR-V7↓[44]
Transgenic miceGnetin C
Pterostilbene
R26MTA1; Pten+/f; Cre+GnC-Diet 35 and 70 mg/kg diet, or
Pter-Diet 70 mg/kg diet
Reduction of cell proliferation, angiogenesis and inflammation;
MTA1↓, pAkt/Akt↓, PTEN↑,
IL2↓ in serum
[54]
Gnetin CR26MTA1; Ptenf/f, Cre+7 mg/kg bw, i.p., 12 weeksInhibition of cell cycle progression, proliferation and angiogenesis, induction of apoptosis;
MTA1↓, Cyclin D1↓,
p-mTOR/pS6K/p4EBP1↓,
IL2↓ in serum
[53]
↑ upregulation; ↓ downregulation; GnC, Gnetin C; Enz, enzalutamide. DU145 (AR-negative); PC3M (AR-negative): 22Rv1 (AR-positive, AR-V7); VCaP (AR-positive, AR-V7).
Table 3. Gnetin C and other effects.
Table 3. Gnetin C and other effects.
CompoundModelDoseCell/AnimalMechanism of ActionRef.
CardioprotectiveGnetin C
Resveratrol
Other stilbenes
Ex vivo500 µMHuman platelet-collagen adhesion assayRes inhibits arachidonic acid- and thrombin-induced platelet aggregation; Gnetin C more potently inhibits platelet–collagen adhesion compared to Res[63]
Gnetin C
MSE
Resveratrol
Grape extract
In vitro0.05 nM angiotensin II + GnC 30 µM, MSE 300 µg/mL or Res 30 µMRadioligand binding assay in transfected HEK-293 cells transfected COS-1 cellsGnC and MSE: inhibit ATII-type 1 receptor binding;
GnC, MSE, grape extract, Res: mild agonists at PPARα and PPARγ
[21]
NeuroprotectiveGnetin C
Resveratrol
ε-viniferin
In vitro0–20 µMSH-SY5Y cellsGnetin C more potently Aβ42↓ secretion, BACE1↓, Aβ oligomers↓, Aβ monomers↑, MMP-14↑, mitigated Aβ42-induced cytotoxicity[64]
Gnetin CIn vitro0–10 µMU373MG and SH-SY5Y cells: poly-IC-induced TLR3-mediated inflammationIFN-β↓, STAT1 phosphorylation↓, CCL2↓, CCL5↓[65]
MetabolicGnetin C
MSE
Resveratrol
In vivoHFCD diet supplemented with 0.5% MSE, 12 wkHFCD diet-induced NAFLD mouse modelMSE: body weight↓, liver weight↓, Tg↓, NEFAs↓, ALT↓, liver steatosis↓, hepatic fibrosis↓[66]
In vivoHFCD diet supplemented with GnC or Resv 150 mg/kgbw/day, 12 wkHFCD diet-induced NAFLD mouse modelGnC and Res: body weight↓, liver weight↓, IL-1b↓, adiponectin↑, liver steatosis↓, hepatic fibrosis↓, collagen deposition↓, COL1A1↓, TGFβ1↓;
GnC: glucose↓, insulin sensitivity↑, lipids↓, ACC1↓, chREBP↓, DGAT1↓, DGAT2↓, MTP↓, LDLR↓, PPARα↓, PGC-1α↓, SIRT1↓
Gnetin C
MSE
In vivo2% MSE-supplemented diet, 21 daysStreptozotocin-induced diabetic mice (model for endothelial senescence)SA-β-gal-positive cells↓, aortic SIRT1↑;
Plasma Gnetin C component 6-fold higher than resveratrol
[16]
Resveratrol
Gnemonosides A, D
In vitro100 µmol/LHUVEC H2O2-induced endothelial senescenceOnly resveratrol component able to SA-β-gal-positive cells↓, SIRT1↑, eNOS↑, PAI1↓
Gnetin C
Resveratrol
Gnetin L
Gnemonosides A, C, D
In vitroConstituents extracted from dried melinjo endosperm and purifiedDPPH radical scavenging activityGnetin C ED50: 10.7 µM
Resveratrol ED50: 13.2 µM
[15]
Pancreatic digestive enzymesGnetin C has greater lipase and α-amylase inhibition than resveratrol
FoodModerate antimicrobial activity
Anti-inflammatory—Anti-agingGnetin C
Resveratrol
In vitro2–8 µMMurine B16 cellsGnC and Res: Similar inhibitory potency of tyrosine activity and melanin biosynthesis; neither is cytotoxic[27]
In vitro2–16 µMCell-free inhibition of tyrosinase enzymeGnC has less direct inhibition of tyrosine activity
Gnetin C
Resveratrol
In vivo10 mg/kg i.p. daily for 7–8 daysLigature-induced periodontitis mouse modelGnC and Res: 8-OHdG↓;
GnC: bone healing↑ and IL-1β↓
[67,68]
In vivoAs aboveNrf2−/− transgenic miceNeither treatment able to induce bone healing
↑ upregulation; ↓ downregulation; Res, resveratrol; MSE, melinjo seed extract; GnC, Gnetin C.
Table 4. Human clinical trials with Gnetin C or MSE.
Table 4. Human clinical trials with Gnetin C or MSE.
Trial TypeTreatmentPopulationNumber of Participants and DurationMarkersOutcomeRef
Randomized, double-blind, placebo-controlledGnetin C 150 mg/day, orallyHealthy Japanese subjectsN = 12
Days: 14
No change in CRP, Tg, 8-OHdG or pentosidine;
LDL-C↓, HDL-C↓, adiponectin↓, NK cells↑
Safety;
Cardioprotective effect
[24]
Randomized, double-blind, placebo-controlledMSE 750 mg daily, orallyNonobese Japanese males,
35–70 yrs old
N = 30
Weeks: 8
Serum uric acid↓, LDL-C no change, HDL-C↑Cardioprotective[21]
No placeboSingle-dose study: Res 6.80 mg/day;
MSE 1000 mg/day, orally
Healthy volunteersN = 10 (6 men, 4 women)
23–34 yrs old
Days: 28
Safety; Pharmacokinetics[22]
Placebo-controlledRepeated doses
MSE 1000 mg,
2000 mg or 5000 mg
Healthy volunteersN = 44 (22 men, 22 women)
32–49 yrs old
Days: 14 and 28
Blood pressure, pulse, body mass index;
biochemical parameters in blood, urine
No placeboMSE tablets (38.5% MSE powder equivalent to 262 mg Gnetin C):
20 tabs/day
Healthy volunteersN = 5 (3 males, 2 females)
34–46 yrs old
Days: 28
Blood, circulating:
Immune cells
Surface immune receptors
Treg cells
CTL (GZMB)
NK (NKG2D receptor)
Inflammatory cytokines IFNγ; TNFα
8-OHdG↓
Safety
Antioxidant effects
Effects on circulating lymphocytes
Chemopreventive potential
[23]
Randomized, double-blind, placebo-controlledMSE 150 mg or 300 mg daily vs. placeboHealthy young volunteersN = 42
Days:14
HMW/total APN↑
LDL-C↓, ALT↓
Anti-inflammatory
Insulin sensitivity
Cardioprotective
[25]
↑ upregulation; ↓ downregulation; MSE, melinjo seed extract; Res, resveratrol.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Campanelli, G.; Levenson, A.S. Gnetin C in Cancer and Other Diseases: What Do We Know So Far? Nutrients 2025, 17, 863. https://doi.org/10.3390/nu17050863

AMA Style

Campanelli G, Levenson AS. Gnetin C in Cancer and Other Diseases: What Do We Know So Far? Nutrients. 2025; 17(5):863. https://doi.org/10.3390/nu17050863

Chicago/Turabian Style

Campanelli, Gisella, and Anait S. Levenson. 2025. "Gnetin C in Cancer and Other Diseases: What Do We Know So Far?" Nutrients 17, no. 5: 863. https://doi.org/10.3390/nu17050863

APA Style

Campanelli, G., & Levenson, A. S. (2025). Gnetin C in Cancer and Other Diseases: What Do We Know So Far? Nutrients, 17(5), 863. https://doi.org/10.3390/nu17050863

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