Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 May 1.
Published in final edited form as: Semin Cancer Biol. 2020 Feb 29;80:107–117. doi: 10.1016/j.semcancer.2020.02.012

Metastasis-associated protein 1-mediated antitumor and anticancer activity of dietary stilbenes for prostate cancer chemoprevention and therapy

Anait S Levenson 1,*
PMCID: PMC7483334  NIHMSID: NIHMS1572042  PMID: 32126261

Abstract

Dietary bioactive polyphenols that demonstrate beneficial biological functions including antioxidant, anti-inflammatory, and anticancer activity hold immense promise as effective and safe chemopreventive and chemosensitizing natural anticancer agents. The underlying molecular mechanisms of polyphenols’ multiple effects are complex and these molecules are considered promising targets for chemoprevention and therapy. However, the development of novel personalized targeted chemopreventive and therapeutic strategies is essential for successful therapeutic outcomes. In this review, we highlight the potential of metastasis-associated protein 1 (MTA1)-targeted anticancer and antitumor effects of three dietary stilbenes, namely resveratrol, pterostilbene, and gnetin C, for prostate cancer management. MTA1, an epigenetic reader and master transcriptional regulator, plays a key role in all stages of prostate cancer progression and metastasis. Stilbenes inhibit MTA1 expression, disrupt the MTA1/histone deacetylase complex, modulate MTA1-associated Epi-miRNAs and reduce MTA1-dependent inflammation, cell survival, and metastasis in prostate cancer in vitro and in vivo. Overall, the MTA1-targeted strategies involving dietary stilbenes may be valuable for effective chemoprevention in selected subpopulations of early stage prostate cancer patients and for combinatorial strategies with conventional chemotherapeutic drugs against advanced metastatic prostate cancer.

Keywords: MTA1, prostate cancer, natural products, dietary stilbenes, resveratrol, pterostilbene, gnetin C, chemoprevention, therapeutics, pharmacological targeting

1. Introduction

Natural products offer promising options to reduce the progress of cancer. In fact, most chemotherapeutics currently in use are derived from natural sources, which continue to provide key scaffolds for drug development [1]. Natural products mainly consist of bioactive polyphenols present in vegetables, fruits, or seeds. Polyphenols’s chemical composition varies from simple to highly polymerized structures containing multiple phenol rings [2, 3]. Different classes of polyphenols include flavonoids, phenolic acids, lignans, and stilbenes. Polyphenols are known to exert antioxidant, anti-inflammatory, and anticancer effects in in vitro and in vivo experimental settings through pleotropic mechanisms affecting various signaling pathways and cell functions [4, 5]. The potential mechanisms of action include phenol groups’ nonspecific scavenging abilities as well as specific interactions based on the particular structural and conformational properties of select polyphenols and the biological target. Importantly, there is epidemiological evidence for an association between polyphenol intake and low cancer risk [6].

Prostate cancer is the second most commonly diagnosed cancer and the second leading cause of cancer death among men in developed countries [7, 8]. The occurrence of prostate cancer is increasing because of population aging, the primary risk factor for slow- growing prostate cancer. Prostate cancer is unique in that if a man lives long enough he could develop some form of the disease that may never progress. On the other hand, due to other risk factors that include family history, ethnicity and Western diet [9, 10], men may develop aggressive prostate cancer that requires treatment. Treating patients who have developed high-grade localized prostate cancer can have a survival benefit but usually is associated with toxicity and significant adverse effects. Understandably, an interest in cost-effective and nontoxic nutritional chemopreventive measures is rising with the confidence that a substantial portion of prostate cancer cases could be prevented by applying effective “prostate cancer-specific diets” that contain bioactive dietary polyphenols and micronutrients [11, 12]. Indeed, epidemiological data and certain case-control studies have found that high consumption of vegetables, fruits, or soy foods was associated with reduced prostate cancer risk [1318]. Dietary strategies targeting various mechanisms that are associated with prostate cancer progression also have been suggested [1921].

A large number of cellular and molecular targets have been identified that could be involved in the cytotoxic and anticancer actions of dietary polyphenols. A combination of phytochemicals rather than any single polyphenol is likely responsible for any health benefits. Even so, it is vital to identify specific molecular targets of a particular class of polyphenols for a given type of cancer to develop targeted precision chemopreventive and therapeutics strategies. The objective of the current review is to present evidence about the role and potential mechanism of the action of dietary stilbenes in the inhibition of epigenetic modifier metastasis-associated protein 1 (MTA1) and MTA1-associated cell signaling, tumor progression, and metastasis in prostate cancer. Identifying the oncogenic MTA1 as a molecular target for stilbenes offers a novel advantage of stilbene-rich diets for chemoprevention as well as for effective combined strategy with approved drugs against advanced metastatic prostate cancer.

2. Stilbenes: Resveratrol, Pterostilbene, and Gnetin C

Stilbenes are known to possess a wide range of biological activities including antioxidative, anti-inflammatory, antifungal, antibacterial, antiviral, and antitumor activities. Readers who are interested in a detailed discussion of these effects are directed to several excellent articles and book chapters that cover those topics [3, 2231]. Stilbenes have a common C6-C2-C6 structure, consisting of two aromatic rings linked through a two-carbon bridge with a double bond. Stilbenes are produced by certain plants in response to different environmental stressors such as UV light or infections [3234]. The parent compound of this family is resveratrol, which occurs as a monomer and as oligomers [23, 34, 35]. Other stilbenes such as pterostilbene, piceatannol, or trimethoxy-resveratrol are structurally related to resveratrol with the same backbone but differing in the type, number, and position of substituents [36].

Major dietary sources of stilbenes include grapes, wine, blueberries, peanuts, Itadori tea common in China and Japan, and edible melinjo plant that is used in Southeast Asian and western Pacific Islands’s cuisines [23, 33, 37].

We herein focus on the targeted antitumor and anticancer activities of three stilbenes: resveratrol, pterostilbene, and gnetin C. Resveratrol (3,5,4′-trihydroxy-trans-stilbene) is one of the best characterized stilbenes, found mostly in grapes, wine, and peanuts. Pterostilbene (3,5-dimethoxy-4′-hydroxy-trans-stilbene) is a natural methoxylated analog of resveratrol, found mostly in blueberries and grapes. Gnetin C is a dimer-resveratrol (5-{(2S,3S)-4-hydroxy-2-(4-hydroxyphenyl)-6-[(E)-2-(4-hydroxyphenyl)vinyl]-2,3-dihydro-1-benzofuran-3-yl}−1,3-benzenediol) , found in the melinjo plant Gnetum gnemon (Fig. 1).

Figure 1.

Figure 1.

Chemical structures of resveratrol, pterostilbene, and gnetin C.

3. MTA1, MTA1/nucleosome remodeling and deacetylase (NuRD) complex, and cancer

MTA1 belongs to the MTA family of cancer progression-related proteins that are part of the NuRD co-repressor complex, which is involved in chromatin remodeling via histone deacetylation and gene-specific transcriptional regulation [3840]. The MTA family includes three major proteins (MTA1, MTA2, and MTA3) and several isoforms, which are engaged in numerous functions of NuRD complex. They interact with a variety of chromatin remodeling factors and enzymes leading to nucleosome remodeling and transcriptional repression or activation of target genes [41, 42]. MTA1 itself does not have intrinsic enzymatic activity and has not been shown to directly interact with DNA [40, 43, 44]. In cancer, MTA1 can interact with histone deacetylase (HDAC 1/2) complex but may also perform histone-independent functions including deacetylation of non-histone proteins [41, 4548]. In addition, MTA1 regulates the expression of target genes through NuRD-independent mechanisms [40].

MTA1’s structural organization includes six functional modular domains by which the functions of MTA1 were extrapolated (Fig. 2). The functional significance of the BAH, ELM, and SANT domains were hypothesized from studies of proteins containing similar homologous domains [4951]. Although experimental evidence in support of direct MTA1’s DNA-binding activity is lacking, a GATA-like zing-finger domain commonly found in the GATA transcription factors [52, 53] suggests that MTA1 may be a bona fide transcription factor. MTA1 also contains one monopartite and two bipartite nuclear localization signals (NLSs) in addition to a proline-rich Src-homology domain, which is essential for interacting with other proteins [43, 54].

Figure 2.

Figure 2.

Schematic representation of the MTA1 protein structure and correlated putative functions. BAH, bromo-adjacent homology; ELM, egl-27 and MTA1 homology; SANT, SWI, ADA2, N-CoR, TFIIIB-B (shares homology with DNA-binding domain of Myb-related proteins); ZnF, zing finger motif (GATA-type); NLS, nuclear localization signals; BP1 and BP2, bipartite NLS; SH, src-homology binding motifs (proline-rich). aa: amino acids

MTA1 is highly expressed in various cancers and both nuclear and cytoplasmic accumulation of MTA1 has been reported [47, 5557]. In human prostate cancer, immunohistochemical staining has documented predominantly nuclear staining associated with higher Gleason scores and metastasis [42, 58, 59], which explains MTA1’s epigenomic and genomic functions. On the other hand, MTA1 localization in the cytoplasm suggests possible nongenomic functions for MTA1 in prostate cancer (yet to be discovered), analogous to those reported in breast cancer that are a hyperstimulation of extranuclear activities of the estrogen receptor α (ERα) [60, 61]. MTA1 has been also detected in normal prostates, bladders, and brains isolated from wild-type (wt) mice with the highest MTA1 expression in testes [41, 42]. In prostate-specific Pten (phosphatase and tensin homolog) - loss mouse models, MTA1 was detected in both prostate epithelial glands and reactive stroma compartments, indicating its role in inflammation-driven processes [62]. The role of MTA1 in normal prostates remains unknown.

4. MTA1’s clinical significance and functional role in prostate cancer progression and metastasis

The clinical significance of MTA1 in human prostate cancer has been revealed [58, 59, 63, 64]. MTA1 is expressed in early stage prostate cancer but its overexpression is closely associated with the clinico-pathological characteristics of prostate cancer progression: high Gleason scores, development of castrate resistance, recurrence, and metastasis. Amplification of Mta1 gene was detected in neuroendocrine prostate cancer [65, 66]. MTA1 plays a special role in prostate cancer bone metastasis [58, 59, 67]. We found a strong association between high MTA1 levels and disease recurrence and metastasis, particularly in African American patients, which suggested to us MTA1’s possible race-specific driver role of aggressive prostate cancer progression and metastasis [59]. These observations warrant further studies for a better understanding of the specific role of MTA1 in each stage of prostate cancer and its potential value as a prognostic and predictive biomarker as well as chemopreventive and/or therapeutic target for prostate cancer progression and metastasis.

Almost a decade ago, my group identified MTA1 as a part of the “bone metastatic signature” in an experimental setting where we incubated various prostate cancer cells, namely LNCaP, C4–2, DU145, PC3, and PCa2b lines with bone chunks isolated from rats’ tibias [58]. Analysis of differential gene expression profiling revealed MTA1 as a novel putative component of the bone metastatic “vicious cycle”, which suggested MTA1’s explicit role in prostate cancer bone-specific metastasis. Although we proceeded to detect striking nuclear overexpression of MTA1 in bone lesions of human metastatic prostate cancer samples [58, 59], only recently were we able to demonstrate the functional role of MTA1 in the formation of bone metastasis using an experimental bone metastatic model in prostate cancer xenografts [67]. We found that MTA1 silencing (shMTA1) in PC3M prostate cancer cells impaired tumor growth and reduced colonization and development of bone metastasis in subcutaneous and intracardiac prostate cancer xenografts, respectively. Moreover, we demonstrated the critical role of MTA1 as an upstream regulator of Cathepsin B, a cysteine protease actively involved in prostate cancer bone metastasis [6769]. MTA1’s role is not exclusive to bone metastasis since we detected overexpression of MTA1 in other metastatic lesions obtained from prostate cancer patients [59]. This was expected, since the active involvement of MTA1 in two major biological processes such as epithelial-to-mesenchymal transition (EMT) and angiogenesis, which are characteristics of prostate cancer progression and metastasis, is well documented. The role of MTA1 as an inducer and promoter of EMT pathways and metastatic progression has been documented in many cancers [42, 7073]. In prostate cancer, mechanistic studies with loss-of-function and overexpression of MTA1 have demonstrated MTA1’s involvement in the malignant phenotype of cells via pAkt/E-cadherin pathway [74]. E-cadherin, in turn, regulates MTA1-dependent tumor cell adhesion, mobility, invasion and cellular polarity [75]. We detected an inverse correlation between MTA1 and E-cadherin in prostate tissues of prostate-specific Pten knockout mice (Pb-Cre4; Ptenf/f), in which increased levels of MTA1 were concomitant with decreased levels of E-cadherin and increased levels of vimentin [62]. We also have reported modulation of EMT markers upon MTA1 knockdown in LNCaP and DU145 prostate cancer cells [62]. Further, MTA1 silencing led to a significant increase in the E-cadherin mRNA and protein levels in PC3M cells and PC3M-xenograft tumor tissues [67]. One additional mechanism, by which MTA1 regulates E-cadherin, involves activation of MTA1-associated microRNAs [76] (see section 6.3).

The pro-angiogenic character of MTA1 has been well documented in various cancers [47, 48, 7781]. One early observation about the link between MTA1 and angiogenesis was that the hypoxia inducible factor 1α (HIF-1α) is regulated by a MTA1/HDAC1 complex, which stabilizes and transcriptionally activates HIF-1α [48]. Reports demonstrated that MTA1 is overexpressed under hypoxic and in certain treatment conditions, physically interacts with HIF-1α causing its deacetylation and stabilization [47, 48, 81]. Moreover, there is a positive correlation between high MTA1 levels and transcriptional activation of HIF-1α leading to enhanced expression of vascular endothelial growth factor (VEGF), a target molecule of HIF-1α [47, 80]. In addition to HIF-1α and VEGF, there are other angiogenic factors such as angiopoietin1/2, cyclooxygenase 2 (COX-2), matrix metalloproteinases (MMPs), transforming growth factor β (TGFβ), and interleukins (IL6, IL8, and IL-1β) that are involved in prostate cancer angiogenesis [82]. We have shown the contribution of MTA1 in tumor neovascularization and in HIF-1α-, VEGF-, IL-1β-induced angiogenesis in prostate cancer in vitro and in vivo. In cultured MTA1 knockdown PC3/PC3M prostate cancer cells, VEGF levels in conditioned media and HIF1-α mRNA and protein levels in cell extracts were significantly decreased [58, 83]. Moreover, MTA1 silencing considerably reduced the ability of PC3 cells to induce endothelial cell chemotaxis in migration assay [58]. Furthermore, LNCaP and DU145 MTA1 knockdown cells showed decreased levels of IL-1β, a known pro-angiogenic factor that potentiates VEGF expression [62, 82]. All three of these angiogenic factors, i.e. HIF-1α, VEGF-C, and IL-1β, are directly regulated by MTA1, according to our differential MTA1 chromatin IP based deep sequencing (ChIP-Seq) data, in which tracks for each gene loci showed decreased MTA1 occupancy of respective promoters in MTA1 reduced prostate tissues [62, 83]. Most importantly, we demonstrated the link between MTA1 and tumor angiogenesis in vivo [58, 62, 83, 84]. Subcutaneous tumors from LNCaP and DU145 MTA1 knockdown xenografts showed decreased VEGF staining and a reduction in microvessel density (MVD) assessed by CD31 staining of blood vessel’s endothelial cells, compared to control xenografts expressing high levels of MTA1 [58]. In DU145 MTA1 knockdown orthotopic xenografts, tumors showed significant reduction in CD31-positive areas accompanied by lesser metastasis and in fewer organs [84]. In contrast, mice prostate tissues from autochthonous prostate cancer models, namely prostate-specific Pten heterozygous (Pb-Cre+; Pten+/f;) and Pten knockout (Pb-Cre+; Pten f/f;) mice, which express high levels of MTA1, exhibit higher levels of pro-angiogenic factors TGFβ, HSP90, and IL-1β [62] and high levels of circulating VEGF-C and IL-1β detected in mouse sera [83]. Likewise, prostate tissues from prostate-specific MTA1 transgenic mice, MTA1-tg, (Pb-Cre+; Rosa26+/MTA1 and Pb-Cre+; Rosa26MTA/MTA1), which express high levels of MTA1 showed significantly higher levels of HIF-1α at both mRNA and protein levels compared to normal prostates [83]. Collectively, this data demonstrate MTA1’s active involvement in prostate cancer angiogenesis through the regulation of pro-angiogenic factors.

While current evidence in the literature links MTA1 to processes associated with cancer metastasis, our studies suggest that MTA1 is involved in almost all stages of prostate cancer progression [42]. Prostate cancer is associated with inflammation, which is characteristic of an accepted precursor to prostate cancer, prostatic intraepithelial neoplasia (PIN) [85]. Levels of MTA1 were significantly higher in the prostate tissues from prostate-specific Pten heterozygous mice with high grade PIN characterized by the presence of reactive stroma compared to normal prostate, suggesting that MTA1 may be involved in the early inflammation stage of prostate cancer. In cancer-prone prostate-specific Pten knockout model, MTA1 expression was dramatically higher and was associated with the activation and promotion of oncogenic and survival pathways [62]. Mechanistically, it has been shown that MTA1 either regulates or is regulated by several key molecules that have been implicated in the induction and activation of reactive stroma leading to inflammation and tumor promotion. The link between MTA1 and NF-κB, TGFβ, and c-Myc has been demonstrated in various cancers [8690]. In the prostate-specific Pten loss models of prostate cancer, high levels of MTA1 were accompanied with increased levels of AR, TGFβ, NF-κB (p65), IL-1β, HSP90, Akt/pAkt, Cyclin D1, Notch2, and ETS2 [62]. We also detected a reduction in the levels of Cyclin D1, c-Myc, pAkt/Akt, ETS2, Notch2, IL-1β, and HSP90 in MTA1 knockdown prostate cancer cell lines at both mRNA and protein levels confirming transcriptional regulation of these genes by MTA1 (MTA1 ChIP-Seq analysis) [62]. Therefore, MTA1, which is expressed in both prostate tumor and reactive stroma, has pro-inflammatory, pro-survival, pro-angiogenic and metastatic roles in Pten loss-driven prostate tumor growth and progression. Of a note, our MTA1-tg mice developed high grade PIN detected at 14 weeks of age but failed to develop invasive adenocarcinoma at up to 1 year of age, suggesting that MTA1 alone is insufficient to cause prostate cancer (unpublished data, Levenson). Nevertheless, the MTA1-targeted strategies may prove successful as chemopreventive and therapeutic options for early stage and metastatic prostate cancer, respectively.

5. MTA1-targeted chemoprevention and therapeutic potential of dietary stilbenes in prostate cancer

5.1. Targeted therapy in prostate cancer

Although the link between diet and cancer is complex, epidemiological data confirm diet as a risk factor for prostate cancer [91] and indicate a reduced prostate cancer risk associated with red wine consumption, attributable to high resveratrol content [92]. The fact that prostate cancer is an age-related slow-growing disease linked with diet as a risk factor makes prostate cancer the most appropriate for chemoprevention by dietary bioactive polyphenols with anti-inflammatory, antioxidant and anticancer properties. Primary prostate cancer chemoprevention would be particularly valuable for the subclass of patients for whom we usually recommend active surveillance or watchful waiting. These patients are not eligible for surgery or any other treatments due to their early stage, which might or might not progress to prostate cancer. Unfortunately, almost half of these patients develop prostate cancer, posing an urgent demand for developing effective chemoprevention tactics such as a “chemopreventive” diet. Now it became clear that it is imperative to customize chemopreventive strategies by defining the particular subset of patients (active surveillance) likely to respond to certain dietary compounds (dietary stilbenes) via specific signaling pathways (MTA1-signaling). In addition, various combination strategies with dietary stilbenes and chemotherapeutic drugs may show effective therapeutic effects by targeting MTA1 through the same or different signaling pathways. Thus, combinatorial approaches with other MTA1 pharmacological inhibitors should be considered for possible synergistic and minimizing adverse side effects in targeted patient populations with advanced prostate cancer. Table 1 summarizes MTA1-mediated anticancer effects of stilbenes and combinations with synthetic agents in prostate cancer.

Table 1.

Stilbenes’ MTA1-mediated anticancer effects in prostate cancer

Stilbene Cell Type Mouse Model / Route Effects Signaling / Markers Reference #
Resveratrol LNCaP
DU145
Apoptosis⭡ MTA1⭣; HDAC1⭣
p53 activation via acetylation⭡
p21, Bax promoter activation
Bax⭡; BCL-2⭣
p53 acetylation⭡⭡
[46]
Resveratrol + SAHA LNCaP
DU145
Apoptosis⭡⭡
No treatment PC3
PC3-siMTA1
Invasion⭣
Angiogenesis⭣
VEGF⭣ [58]
LNCaP-siMTA1
DU145-siMTA1
s.c xenografts / i.p.
Tumor volume⭣
Tumor weight⭣
Ki67⭣; CD31⭣
Resveratrol
Trimethoxy-resveratrol
Piceatannol
Pterostilbene
LNCaP
DU145
PC3M
Cell viability⭣
Colony formation⭣
[36]
LNCaP-Luc s.c. xenografts / oral gavage Tumor volume⭣ Serum IL-6⭣
Resveratrol
Trimethoxy-resveratrol
Piceatannol
Pterostilbene
LNCaP
DU145
PC3M
MTA1⭣
MTA1⭣⭣
[84]
Pterostilbene DU145-EV
DU145-shMTA1
DU145-Luc orthotopic xenografts
/ i.p.
Tumor volume⭣
Apoptosis⭡
Angiogenesis⭣
Metastasis⭣
MTA1-mediated
p53 acetylation⭡
Ki67⭣; M30⭡; CD31⭣
Resveratrol
Pterostilbene
DU145
22Rv1
DU145-EV
DU145-shMTA1
MTA1/HDAC ½⭣
PTEN acetylation⭡
pAkt/Akt⭣
[45]
Resveratrol DU145-EV
DU145-shMTA1
DU145- Luc Orthotopic xenografts / i.p. Tumor volume⭣
Apoptosis⭡
MTA1⭣; PTEN⭡; pAkt⭣ Ki67⭣
No treatment LNCaP-EV
LNCaP-shMTA1
DU145-EV
DU145-shMTA1
MTA1-mediated
NF-κB⭣; IL-1β⭣
Hsp90⭣; E-cad⭡; Vimentin⭣
c-Myc⭣; CyclinD1⭣; Notch2⭣
ETS2⭣; pAkt/Akt⭣; p21⭡; p27⭡
[62]
Pterostilbene Pb-Cre+; Pten+/f / diet PIN formation⭣
Proliferation⭣
Apoptosis⭡
MTA1⭣; PTEN⭡; SMA⭡; CK8⭡
pAkt/Akt⭣; c-Myc⭣; Cyclin D1⭣
Notch2⭣; TGFβ⭣; ETS2⭣;
Hsp90⭣; AR⭣; p21⭡; p27⭡
Ki67 ⭣; cl caspase 3⭡
Pb-Cre+; Ptenf/f / i.p. Adenocarcinoma⭣
Proliferation⭣
Apoptosis⭡
Angiogenesis⭣
MTA1⭣; pAkt/Akt⭣; AR⭣
E-cad⭡; IL-1β⭣; Hsp90⭣
Ki67⭣; CD31⭣; cl caspase 3⭡
Ac p53⭡; Bak⭡; VEGFc⭣
Pterostilbene
Pterostilbene + SAHA
LNCaP
PC3M
PC3M-NS
PC3M-shMTA1
Cell viability⭡ MTA1-mediated HIF1α⭣
VEGFc⭣; IL-1β⭣
[83]
Pb-Cre+; Ptenf/f; Rosa26Luc/+ / i.p. Hg PIN formation⭣
Apoptosis⭡
Angiogenesis⭣
MTA1/HIF1α⭣; p27⭡
SMA⭡; CK8⭡; Ki67⭣
Cl caspase 3⭡; CD31⭣
Serum VEGFc⭣
Serum IL-1β⭣
Pb-Cre+; Rosa26MTA1/+ MTA1/HIF1α⭡
Resveratrol
Pterostilbene
Gnetin C
DU145
PC3M
DU145-NS
DU145-shMTA1
PC3M-NS
PC3M-shMTA1
Cell viability⭣
Colony formation⭣
Migration⭣
MTA1/ETS2⭣ [105]
No treatment PC3M-NS
PC3M-shMTA1
Proliferation⭣
Colony formation⭣
Migration⭣
Invasion⭣
MTA1/CTSB⭣; E-cad⭡ [67]
PC3M-shMTA1-Luc s.c. xenografts / i.p. PC3M-shMTA1-Luc intracardiac xenografts Tumor growth⭣
Bone metastasis⭣
Ki67⭣
MTA1/CTSB⭣; E-cad⭡
Pb-Cre+; Rosa26MTA1/+ MTA1/CTSB⭡
No treatment LNCaP-EV
LNCaP-shMTA1
MTA1-regulated
Epi-miRNAs
MTA1⭣; Epi-miR-22⭣ E-cad⭡ [76]
Pb-Cre+; Rosa26MTA1/+
Pb-Cre+;
Rosa26MTA1/MTA1
MTA1⭡; Epi-miR-22⭡ E-cad⭣
Resveratrol LNCaP OncomiRs⭣
TS miRs⭡
miR-17~92⭣; miR-106a⭣ miR-106b⭣ [126]
Resveratrol
Pterostilbene
LNCaP
DU145
PTEN⭡
miR-17–5p⭣; miR-20a⭣
miR-106a⭣; miR-106b⭣
[127]
Pterostilbene DU145-EV
DU145-miR-17/106a
DU145-miR- Luc s.c.xenografts / i.p. Tumor growth⭣
Apoptosis⭡
PTEN⭡
Ki67⭣; M30⭡; cl caspase 3⭡
Tumor & serum miR-17–5p⭣
Tumor & serum miR-106a-5p⭣

⭡, upregulation;

⭣, downregulation

5.2. Resveratrol and prostate cancer

The anticancer effects of resveratrol acting through various signaling pathways in prostate cancer have been reported in numerous publications [93100]. Our first report on resveratrol’s inhibition of MTA1/NuRD complex, which restored p53 acetylation/activation causing apoptosis in prostate cancer cells, opened the door for comprehensive studies of MTA1-mediated epigenetic and genetic molecular mechanisms of stilbene compounds’ action in vitro and in vivo [46]. We found that by disrupting the MTA1/HDAC1 unit and deregulating its deacetylation function, resveratrol reverses p53 acetylation, leading to the activation of pro-apoptotic Bax and p21, and ultimately causing apoptosis in DU145 (mut p53) and LNCaP (wt p53) prostate cancer cells. Interestingly, the clinically approved HDAC inhibitor, suberoylanilide hydroxamic acid (SAHA), alone did not have any effect on p53 acetylation but amplified resveratrol-induced increase of the acetylated p53 in both cell lines. Combined treatment with resveratrol and SAHA significantly increased apoptosis [46]. These results suggested the epigenetic reader MTA1 as a novel molecular target of resveratrol and the combination of resveratrol and HDAC inhibitor(s) as a possible effective therapeutic approach against prostate cancer. Due to resveratrol’s known low bioavailability, we began a search for analog(s) with better pharmacokinetic properties and potent biological effects. We compared the ability of six different analogs of resveratrol to inhibit prostate cancer cell proliferation, colony formation, and tumor growth in subcutaneous (s.c.) xenografts and found that some analogs, including pterostilbene, are more potent in inhibiting prostate cancer cells compared to resveratrol [36].

5.3. Pterostilbene and prostate cancer

In contrast to resveratrol, publications on the effects of pterostilbene in prostate cancer are limited [101104]. We found that pterostilbene was the most potent inhibitor of MTA1 in cultured prostate cancer cells and showed MTA1-dependent tumor regression and inhibition of metastasis in orthotopic DU145 xenografts through increased p53 acetylation, higher apoptotic index, and decreased angiogenesis [84]. Resveratrol and pterostilbene also rescued PTEN acetylation through inhibition of MTA1/HDAC 1/2, which leads to lower pAkt levels and inhibition of the Akt survival-signaling pathway [45]. The relevant in vivo efficacy of pterostilbene as a MTA1-targeted chemopreventive and intervention strategy has been demonstrated using prostate-specific Pten loss immunocompetent pre-clinical mouse models [62]. Pten loss driven prostate cancer progression in prostate-specific Pten heterozygous and Pten-null mice bear a close resemblance to the human disease with an intact immune system. Primary chemopreventive design included diet supplementation with 100 mg/kg diet pterostilbene in Pten heterozygous mice that develop PIN at 7–8 months of age. The intervention strategy involved Pten-null mice that mimic stage-defined progression of human prostate cancer, developing characteristics of invasive adenocarcinoma by 15–20 weeks of age. Because Pten loss resulted in a marked increase in MTA1 expression leading to activation of MTA1-dependent survival and metastasis-associated signaling pathways, we found that pterostilbene, both as a dietary supplement and daily injection, caused MTA1-dependent inhibition of inflammation (NF-κB, HSP90, and IL-1β), tumor growth (pAkt/Akt, AR, Cyclin D1, ETS2, TGFβ, c-Myc, and NOTCH2), EMT (E-cadherin, and vimentin), and angiogenesis (VEGF, CD31, and IL-1β). Pterostilbene induced marked apoptosis (Acp53, p21, p27, cleaved caspase 3), resulting in reduction of PIN lesions in chemopreventive experiments and adenocarcinomas in cancer-prone Pten-null mice [62]. We also provided preclinical evidence for the benefits of MTA1-targeted combinatorial pterostilbene with SAHA strategy. Data showed that pterostilbene sensitized tumor cells to SAHA treatment causing additional decline of tumor progression in Pten-null mice. The effects were dependent on the reduction of MTA1/HIF-1α signaling [83].

5.4. Gnetin C and prostate cancer

So far, there are only two publications available for the effects of gnetin C in prostate cancer cells including our own study on MTA1/ETS2-mediated effects of gnetin C [105, 106]. We have recently revealed that gnetin C exhibited potent MTA1 inhibition in prostate cancer cells compared to resveratrol and pterostilbene. In addition, we reported for the first time that gnetin C demonstrated significant MTA1-mediated induction of apoptosis in prostate cancer cells while inhibiting cell viability, colony formation, and migration with more efficacy than resveratrol or pterostilbene. Notably, gnetin C also inhibited oncogenic ETS2 in prostate cancer cells via MTA1-dependent and independent mechanisms [105]. As expected, gnetin C showed potent inhibition of subcutaneous prostate cancer xenograft tumors (unpublished data, Levenson).

6. Main MTA1-mediated mechanisms of stilbenes’ action in prostate cancer

6.1. Stilbenes and MTA1/HDAC complex-associated epigenetic mechanisms

The MTA1-mediated molecular mechanisms that contribute to the chemopreventive and therapeutic capacities of natural stilbenes are many-sided including epigenetic effects associated with the MTA1/HDAC1/2 inhibition leading to posttranslational modifications of tumor suppressors and microRNAs (miRNAs, miRs) modulation [107] (Fig. 3). As an epigenetic reader, MTA1 in complexes with HDAC1 and HDAC2 participates in DNA sliding of NuRD and promotes the deacetylation of histones and certain non-histone proteins [38, 39, 46, 108]. As a “master co-regulator”, MTA1 plays a role in recruiting the protein complexes to the promoters of specific genes for transcriptional activation or silencing [42, 43, 109111]. As we already mentioned, stilbenes exhibit epigenetic actions through disruption of the NuRD co-repressor complex (Fig. 3A). We have shown that both resveratrol and pterostilbene promote acetylation and reactivation of tumor suppressors p53 and PTEN through downregulation of the MTA1/HDACs units of the NuRD complex, which lead to inhibition of survival pathways and induction of apoptosis in prostate cancer cells [45, 46]. Another notable target of the MTA1 and HDAC1 is HIF-1α, a key transcriptional regulator of pro-angiogenic VEGF. It has been shown that due to deacetylation by MTA1/HDAC complex, HIF-1α becomes stabilized and transcriptionally active leading to tumor angiogenesis and MTA1-associated metastasis [47, 48]. We too accumulated data on the tight link between high levels of MTA1 and angiogenesis and demonstrated the MTA1-mediated inhibitory effects of stilbenes on HIF-1α and pro-angiogenic factors VEGF and IL-1β in prostate cancer [58, 62, 83, 84]. Further, studies of combinatorial resveratrol /SAHA or pterostilbene/SAHA approach demonstrated benefits of chemosensitization of cancer cells, associated with more efficacy of treatment [46, 83].

Figure 3.

Figure 3.

Main MTA1-mediated mechanisms of stilbenes’ action in prostate cancer. (A) Through the epigenomic pathway, stilbenes downregulate MTA1, HDAC1, and HDAC2 disrupting NuRD corepressor complex deacetylation function, which leads to changes in chromatin conformation. Histone acetylation defines the “open” chromatin landscape, which is characterized by transcriptionally active DNA sequences that serve as docking sites for transcription factors and other effector proteins. Non-histone tumor suppressor proteins p53 and PTEN become acetylated and activated, which leads to inhibition of survival pathways and induction of apoptosis. In contrast, acetylation of HIF-1α causes its destabilization and inactivation resulting in decreased angiogenesis. (B) Through the genomic pathway, stilbenes reverse MTA1’s transcriptional co-repressor or/and co-activator functions. p21, E-cadherin are upregulated by stilbenes in prostate cancer whereas the inhibition of MTA1/ETS2, CyclinD1, and Notch2 is also recorded. (C) Left, Stilbenes inhibit oncogenic miRNAs in prostate cancer: targeting oncomiRs of the miR-17 family restored PTEN expression. Right, MTA1-activated Epi-MiR-22 regulates E-cadherin leading to promotion of EMT and prostate cancer progression.

6.2. Stilbenes and MTA1-associated genomic pathways

A transcriptional link between MTA1 and specific genes such as NF-κB, TGFβ, SMAD7, c-Myc, and PTEN has been shown [89, 90, 112114]. To identify new transcriptional targets of MTA1 and changes in genetic landscape associated with response to pterostilbene, we performed genome-wide analysis by ChIP-Seq using prostate tissue chromatin from prostate-specific Pten loss mice treated or not with pterostilbene (Fig. 3B) [62, 67]. Genes of potential significance were identified by mapping MTA1-binding peaks to RefSeq-annotated transcripts. Along with MTA1 associated genes already reported by others [115117], we prioritized novel candidate MTA1 transcriptional targets in prostate cancer, namely Cathepsin B, E-cadherin, HIF-1α, Cyclin D1, and ETS2 and validated their strong direct or inverse correlation with MTA1 and responsiveness to stilbene treatment in vitro and in vivo [62, 67, 83, 105]. Mechanistic studies such as ChIP and ChIP-re-ChIP experiments will further validate MTA1’s direct involvement in transcriptional regulation of these genes.

6.3. Stilbene- regulated and MTA1- regulated miRNAs

MicroRNAs (miRNAs, miRs) are small non-coding RNAs that play a role in gene regulation [118, 119] and act as oncogenes or tumor suppressors [120]. Aberrant miRNA expression is implicated in prostate cancer and correlates with high Gleason score and clinical recurrence [121123]. Importantly, miRNAs have remarkable stability in serum/plasma, which makes them outstanding “liquid biopsy” biomarkers for cancer diagnosis, prognosis, and therapy response [124, 125]. The first report on resveratrol-regulated miRNAs in prostate cancer using miRNA microarray analysis of LNCaP cells showed significant inhibition of oncogenic miRs and upregulation of tumor suppressor miRs [126]. We further demonstrated that resveratrol and pterostilbene restore PTEN expression in prostate cancer cells by inhibiting miR-17~92, miR-106a~363, and miR-106b~25 clusters, which are members of the miR-17 oncogenic family (Fig. 3C) [127]. Moreover, pterostilbene through downregulation of these miRs in murine tumors and systemic circulation rescued PTEN mRNA and protein levels, leading to reduced tumor growth in xenografts [127]. Accumulating evidence suggests dietary stilbenes as attractive miRNA-mediated chemopreventive and therapeutic agents, and circulating miRNAs as potential chemopreventive and predictive biomarkers for clinical development in cancer [25, 128]

Of particular interest in understanding the MTA1-associated epigenetic network is the area of regulatory miRs that are associated with MTA1. MiRNAs that are regulated by or are regulating epigenetic factors are called “Epi-miRs” [120, 129]. Specifically, HDAC and HAT inhibitors regulate mature miRNAs [130, 131]. We identified Epi-miRNAs regulated by MTA1 using miRNA microarrays differential profiling of LNCaP MTA1 knockdown cells [76]. Among Epi-miRNAs induced by MTA1, we identified miR-22 and demonstrated its direct targeting of the 3′-untranslated region (3ˊUTR) of E-cadherin, mechanistically presenting the MTA1/Epi-miR-22/E-cadherin axis as a novel epigenetic signaling pathway that promotes tumor invasion in prostate cancer (Fig. 3C, right). MTA1-induced overexpression of miR-22 diminished E-cadherin expression resulting in increased cell invasiveness and migration in prostate cancer. Further, we also found positive correlation between MTA1 and miR-22 expression and their inverse correlation with E-cadherin in human prostate cancer samples [76]. The regulation of MTA1/miR-22/E-cadherin axis by stilbenes warrants further investigation to validate Epi-miR-22 as a predictive biomarker and potential therapeutic target in advanced prostate cancer. In addition, miRNAs that target MTA1 have been identified as well [132134] and it would be interesting to test the responsiveness of those miRNAs to stilbenes.

In summary, we suggest MTA1 and MTA1-associated signaling pathways targeting stilbenes as a novel epigenetic personalized chemopreventive and therapeutic strategy to be used either alone or in combination with Food and Drug Administration-approved anticancer drugs for prostate cancer management.

7. Conclusions and perspectives

This review highlighted the potential of dietary stilbenes, namely resveratrol, pterostilbene and dimer-resveratrol gnetin C, in the context of MTA1-targeted chemoprevention and therapy in prostate cancer. Although all three stilbenes target MTA1, they do it with different potencies with gnetin C being the most potent. Studies have reported pterostilbene’s and gnetin C’s superior pharmacokinetics compared to resveratrol [135140], which may explain the observed more potent biological activity of pterostilbene and gnetin C over resveratrol [25, 105]. Notably, we detected accrual of pterostilbene in the murine prostate tissues in Pten loss mice treated with pterostilbene indicating that pterostilbene reaches the target organ [62].

The precise mechanism by which stilbenes regulate MTA1 expression is not completely elucidated and more research should focus on understanding the complex mechanisms of stilbene interaction with MTA1. Nevertheless, current findings on potent inhibition of MTA1 justifies its exploitation as a chemopreventive and therapeutic target in prostate cancer.

Clinical studies in humans have been conducted with all three stilbenes showing their safety [138, 139, 141, 142], however, no human clinical trial has been conducted to evaluate the effects of resveratrol or pterostilbene or gnetin C specifically in prostate cancer. At the time this review was submitted, there was only one somewhat relevant reported trial, in which researchers evaluated the effects of high dose resveratrol on the serum levels of androgens, circulating prostate specific antigen levels, and prostate size in middle-aged men with metabolic syndrome and found only lowered serum levels of the androgen precursors [143]. More attempts were made with combination phytotherapy in prostate cancer, concluding that further exploration of combination with other phytochemicals and/or grape skin extracts for convincing clinical proof is necessary [144146]. Evidently, there are number of challenges in the translational success of natural chemopreventive agents including stilbenes [21].

There is a clear need for personalized targeted approaches for dietary chemoprevention and combinatorial strategies of chemosensitization with approved drugs for better outcomes in prostate cancer. Well-designed human chemoprevention and intervention trials in adequate subpopulation of prostate cancer patients will prove the promise of MTA1-targeted efficient management of prostate cancer.

Acknowledgements

This work is supported by the National Cancer Institute of the National Institutes of Health under Award Number R15CA216070 to AS Levenson. The content is solely the responsibility of the author and does not necessarily represent the official views of the National Institutes of Health. I thank my colleagues, postdoctoral fellows, and students in the Levenson laboratory for all their valuable contribution to our current understanding of the potential of stilbenes in chemoprevention and therapy for prostate cancer. I am grateful to Elena V. Levenson for critically editing the manuscript.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of interest statement

I declare no conflict of interest with the publication of this manuscript.

References

  • [1].Newman DJ, Cragg GM, Natural Products as Sources of New Drugs from 1981 to 2014, J Nat Prod 79 (3) (2016) 629–661. [DOI] [PubMed] [Google Scholar]
  • [2].Martini D, Del Bo C, Porrini M, Ciappellano S, Riso P, Role of polyphenols and polyphenol-rich foods in the modulation of PON1 activity and expression, J Nutr Biochem 48 (2017) 1–8. [DOI] [PubMed] [Google Scholar]
  • [3].Tresserra-Rimbau A, Lamuela-Raventos RM, Moreno JJ, Polyphenols, food and pharma. Current knowledge and directions for future research, Biochem Pharmacol 156 (2018) 186–195. [DOI] [PubMed] [Google Scholar]
  • [4].Demirel Sezer E, Oktay LM, Karadadas E, Memmedov H, Selvi Gunel N, Sozmen E, Assessing Anticancer Potential of Blueberry Flavonoids, Quercetin, Kaempferol, and Gentisic Acid, Through Oxidative Stress and Apoptosis Parameters on HCT-116 Cells, J Med Food (2019) [DOI] [PubMed] [Google Scholar]
  • [5].Xing L, Zhang H, Qi R, Tsao R, Mine Y, Recent Advances in the Understanding of the Health Benefits and Molecular Mechanisms Associated with Green Tea Polyphenols, J Agric Food Chem 67 (4) (2019) 1029–1043. [DOI] [PubMed] [Google Scholar]
  • [6].Zamora-Ros R, Barupal DK, Rothwell JA, Jenab M, Fedirko V, Romieu I, Aleksandrova K, Overvad K, Kyro C, Tjonneland A, Affret A, His M, Boutron-Ruault MC, Katzke V, Kuhn T, Boeing H, Trichopoulou A, Naska A, Kritikou M, Saieva C, Agnoli C, Santucci de Magistris M, Tumino R, Fasanelli F, Weiderpass E, Skeie G, Merino S, Jakszyn P, Sanchez MJ, Dorronsoro M, Navarro C, Ardanaz E, Sonestedt E, Ericson U, Maria Nilsson L, Boden S, Bueno-de-Mesquita HB, Peeters PH, Perez-Cornago A, Wareham NJ, Khaw KT, Freisling H, Cross AJ, Riboli E, Scalbert A, Dietary flavonoid intake and colorectal cancer risk in the European prospective investigation into cancer and nutrition (EPIC) cohort, Int J Cancer 140 (8) (2017) 1836–1844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, Global cancer statistics, 2012, CA Cancer J Clin 65 (2) (2015) 87–108. [DOI] [PubMed] [Google Scholar]
  • [8].Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries, CA Cancer J Clin 68 (6) (2018) 394–424. [DOI] [PubMed] [Google Scholar]
  • [9].Bellamri M, Turesky RJ, Dietary Carcinogens and DNA Adducts in Prostate Cancer, Adv Exp Med Biol 1210 (2019) 29–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Bostwick DG, Burke HB, Djakiew D, Euling S, Ho SM, Landolph J, Morrison H, Sonawane B, Shifflett T, Waters DJ, Timms B, Human prostate cancer risk factors, Cancer 101 (10 Suppl) (2004) 2371–2490. [DOI] [PubMed] [Google Scholar]
  • [11].Mokbel K, Wazir U, Mokbel K, Chemoprevention of Prostate Cancer by Natural Agents: Evidence from Molecular and Epidemiological Studies, Anticancer Res 39 (10) (2019) 5231–5259. [DOI] [PubMed] [Google Scholar]
  • [12].Pejcic T, Tosti T, Dzamic Z, Gasic U, Vuksanovic A, Dolicanin Z, Tesic Z, The Polyphenols as Potential Agents in Prevention and Therapy of Prostate Diseases, Molecules 24 (21) (2019) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Deneo-Pellegrini H, De Stefani E, Ronco A, Mendilaharsu M, Foods, nutrients and prostate cancer: a case-control study in Uruguay, Br J Cancer 80 (3–4) (1999) 591–597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Hebert JR, Hurley TG, Olendzki BC, Teas J, Ma Y, Hampl JS, Nutritional and socioeconomic factors in relation to prostate cancer mortality: a cross-national study, J Natl Cancer Inst 90 (21) (1998) 1637–1647. [DOI] [PubMed] [Google Scholar]
  • [15].Cohen JH, Kristal AR, Stanford JL, Fruit and vegetable intakes and prostate cancer risk, J Natl Cancer Inst 92 (1) (2000) 61–68. [DOI] [PubMed] [Google Scholar]
  • [16].Jain MG, Hislop GT, Howe GR, Ghadirian P, Plant foods, antioxidants, and prostate cancer risk: findings from case-control studies in Canada, Nutr Cancer 34 (2) (1999) 173–184. [DOI] [PubMed] [Google Scholar]
  • [17].Kolonel LN, Hankin JH, Whittemore AS, Wu AH, Gallagher RP, Wilkens LR, John EM, Howe GR, Dreon DM, West DW, Paffenbarger RS Jr., Vegetables, fruits, legumes and prostate cancer: a multiethnic case-control study, Cancer Epidemiol Biomarkers Prev 9 (8) (2000) 795–804. [PubMed] [Google Scholar]
  • [18].Lee MM, Gomez SL, Chang JS, Wey M, Wang RT, Hsing AW, Soy and isoflavone consumption in relation to prostate cancer risk in China, Cancer Epidemiol Biomarkers Prev 12 (7) (2003) 665–668. [PubMed] [Google Scholar]
  • [19].Kaiser A, Haskins C, Siddiqui MM, Hussain A, D’Adamo C, The evolving role of diet in prostate cancer risk and progression, Curr Opin Oncol 31 (3) (2019) 222–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Yedjou CG, Mbemi AT, Noubissi F, Tchounwou SS, Tsabang N, Payton M, Miele L, Tchounwou PB, Prostate Cancer Disparity, Chemoprevention, and Treatment by Specific Medicinal Plants, Nutrients 11 (2) (2019) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Chhabra G, Singh CK, Ndiaye MA, Fedorowicz S, Molot A, Ahmad N, Prostate cancer chemoprevention by natural agents: Clinical evidence and potential implications, Cancer Lett 422 (2018) 9–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Fraga CG, Galleano M, Verstraeten SV, Oteiza PI, Basic biochemical mechanisms behind the health benefits of polyphenols, Mol Aspects Med 31 (6) (2010) 435–445. [DOI] [PubMed] [Google Scholar]
  • [23].Espinoza JL, Inaoka PT, Gnetin-C and other resveratrol oligomers with cancer chemopreventive potential, Ann N Y Acad Sci 1403 (1) (2017) 5–14. [DOI] [PubMed] [Google Scholar]
  • [24].Aggarwal BB, Bhardwaj A, Aggarwal RS, Seeram NP, Shishodia S, Takada Y, Role of resveratrol in prevention and therapy of cancer: preclinical and clinical studies, Anticancer Res 24 (5A) (2004) 2783–2840. [PubMed] [Google Scholar]
  • [25].Kumar A, Levenson AS, Epigenetic mechanisms of resveratrol and its analogs in cancer prevention and treatment. In: Bishayee A, Bhatia D (eds.), Epigenetics of cancer prevention. Elsevier Inc, United Kingdom, (2018) [Google Scholar]
  • [26].Levenson AS, Kumar A, Pterostilbene as a potent chemopreventive agent in cancer. In: Pezzuto JM, Vang O (Eds.), Natural products for chemoprevention: single compounds and combinations. Springer Nature, United Kingdom, (2019) [Google Scholar]
  • [27].Espinoza JL, Elbadry MI, Taniwaki M, Harada K, Trung LQ, Nakagawa N, Takami A, Ishiyama K, Yamauchi T, Takenaka K, Nakao S, The simultaneous inhibition of the mTOR and MAPK pathways with Gnetin-C induces apoptosis in acute myeloid leukemia, Cancer Lett 400 (2017) 127–136. [DOI] [PubMed] [Google Scholar]
  • [28].Kumar A, Butt NA, Levenson AS, Natural epigenetic-modifying molecules in medical therapy. In: Tollefsbol TO (ed.), Medical Epigenetics. Elservier Inc, United Kingdom, (2016) [Google Scholar]
  • [29].Ko JH, Sethi G, Um JY, Shanmugam MK, Arfuso F, Kumar AP, Bishayee A, Ahn KS, The Role of Resveratrol in Cancer Therapy, Int J Mol Sci 18 (12) (2017) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Jiang Z, Chen K, Cheng L, Yan B, Qian W, Cao J, Li J, Wu E, Ma Q, Yang W, Resveratrol and cancer treatment: updates, Ann N Y Acad Sci 1403 (1) (2017) 59–69. [DOI] [PubMed] [Google Scholar]
  • [31].Bishayee A, Cancer prevention and treatment with resveratrol: from rodent studies to clinical trials, Cancer Prev Res (Phila) 2 (5) (2009) 409–418. [DOI] [PubMed] [Google Scholar]
  • [32].Keylor MH, Matsuura BS, Stephenson CR, Chemistry and Biology of Resveratrol-Derived Natural Products, Chem Rev 115 (17) (2015) 8976–9027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Burns J, Yokota T, Ashihara H, Lean ME, Crozier A, Plant foods and herbal sources of resveratrol, J Agric Food Chem 50 (11) (2002) 3337–3340. [DOI] [PubMed] [Google Scholar]
  • [34].Mattivi F, Vrhovsek U, Malacarne G, Masuero D, Zulini L, Stefanini M, Moser C, Velasco R, Guella G, Profiling of resveratrol oligomers, important stress metabolites, accumulating in the leaves of hybrid Vitis vinifera (Merzling x Teroldego) genotypes infected with Plasmopara viticola, J Agric Food Chem 59 (10) (2011) 5364–5375. [DOI] [PubMed] [Google Scholar]
  • [35].Landrault N, Larronde F, Delaunay JC, Castagnino C, Vercauteren J, Merillon JM, Gasc F, Cros G, Teissedre PL, Levels of stilbene oligomers and astilbin in French varietal wines and in grapes during noble rot development, J Agric Food Chem 50 (7) (2002) 2046–2052. [DOI] [PubMed] [Google Scholar]
  • [36].Dias SJ, Li K, Rimando AM, Dhar S, Mizuno CS, Penman AD, Levenson AS, Trimethoxy-resveratrol and piceatannol administered orally suppress and inhibit tumor formation and growth in prostate cancer xenografts, Prostate 73 (11) (2013) 1135–1146. [DOI] [PubMed] [Google Scholar]
  • [37].Rimando AM, Kalt W, Magee JB, Dewey J, Ballington JR, Resveratrol, pterostilbene, and piceatannol in vaccinium berries, J Agric Food Chem 52 (15) (2004) 4713–4719. [DOI] [PubMed] [Google Scholar]
  • [38].Xue Y, Wong J, Moreno GT, Young MK, Cote J, Wang W, NURD, a novel complex with both ATP-dependent chromatin-remodeling and histone deacetylase activities, Mol Cell 2 (6) (1998) 851–861. [DOI] [PubMed] [Google Scholar]
  • [39].Zhang Y, LeRoy G, Seelig HP, Lane WS, Reinberg D, The dermatomyositis-specific autoantigen Mi2 is a component of a complex containing histone deacetylase and nucleosome remodeling activities, Cell 95 (2) (1998) 279–289. [DOI] [PubMed] [Google Scholar]
  • [40].Nair SS, Li DQ, Kumar R, A core chromatin remodeling factor instructs global chromatin signaling through multivalent reading of nucleosome codes, Mol Cell 49 (4) (2013) 704–718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Kumar R, Wang RA, Bagheri-Yarmand R, Emerging roles of MTA family members in human cancers, Semin Oncol 30 (5 Suppl 16) (2003) 30–37. [DOI] [PubMed] [Google Scholar]
  • [42].Levenson AS, Kumar A, Zhang X, MTA family of proteins in prostate cancer: biology, significance, and therapeutic opportunities, Cancer Metastasis Rev 33 (4) (2014) 929–942. [DOI] [PubMed] [Google Scholar]
  • [43].Manavathi B, Kumar R, Metastasis tumor antigens, an emerging family of multifaceted master coregulators, J Biol Chem 282 (3) (2007) 1529–1533. [DOI] [PubMed] [Google Scholar]
  • [44].Kumar R, Wang RA, Structure, expression and functions of MTA genes, Gene 582 (2) (2016) 112–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Dhar S, Kumar A, Li K, Tzivion G, Levenson AS, Resveratrol regulates PTEN/Akt pathway through inhibition of MTA1/HDAC unit of the NuRD complex in prostate cancer, Biochim Biophys Acta 1853 (2) (2015) 265–275. [DOI] [PubMed] [Google Scholar]
  • [46].Kai L, Samuel SK, Levenson AS, Resveratrol enhances p53 acetylation and apoptosis in prostate cancer by inhibiting MTA1/NuRD complex, Int J Cancer 126 (7) (2010) 1538–1548. [DOI] [PubMed] [Google Scholar]
  • [47].Moon HE, Cheon H, Chun KH, Lee SK, Kim YS, Jung BK, Park JA, Kim SH, Jeong JW, Lee MS, Metastasis-associated protein 1 enhances angiogenesis by stabilization of HIF-1alpha, Oncol Rep 16 (4) (2006) 929–935. [PubMed] [Google Scholar]
  • [48].Yoo YG, Kong G, Lee MO, Metastasis-associated protein 1 enhances stability of hypoxia-inducible factor-1alpha protein by recruiting histone deacetylase 1, EMBO J 25 (6) (2006) 1231–1241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Oliver AW, Jones SA, Roe SM, Matthews S, Goodwin GH, Pearl LH, Crystal structure of the proximal BAH domain of the polybromo protein, Biochem J 389 (Pt 3) (2005) 657–664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Grune T, Brzeski J, Eberharter A, Clapier CR, Corona DF, Becker PB, Muller CW, Crystal structure and functional analysis of a nucleosome recognition module of the remodeling factor ISWI, Mol Cell 12 (2) (2003) 449–460. [DOI] [PubMed] [Google Scholar]
  • [51].Manavathi B, Singh K, Kumar R, MTA family of coregulators in nuclear receptor biology and pathology, Nucl Recept Signal 5 (2007) e010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Evans T, Reitman M, Felsenfeld G, An erythrocyte-specific DNA-binding factor recognizes a regulatory sequence common to all chicken globin genes, Proc Natl Acad Sci U S A 85 (16) (1988) 5976–5980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Bresnick EH, Martowicz ML, Pal S, Johnson KD, Developmental control via GATA factor interplay at chromatin domains, J Cell Physiol 205 (1) (2005) 1–9. [DOI] [PubMed] [Google Scholar]
  • [54].Yao YL, Yang WM, The metastasis-associated proteins 1 and 2 form distinct protein complexes with histone deacetylase activity, J Biol Chem 278 (43) (2003) 42560–42568. [DOI] [PubMed] [Google Scholar]
  • [55].Lv ZY, Zhao ZS, Ye ZY, Wang YY, Wang HJ, Yang Q, Metastasis-associated protein 1 (MTA1) in gastric cancer tissues is positively associated with poorer prognosis, Pathol Res Pract 214 (4) (2018) 536–541. [DOI] [PubMed] [Google Scholar]
  • [56].Honjo H, Toh Y, Sohda M, Suzuki S, Kaira K, Kanai Y, Nagamori S, Oyama T, Yokobori T, Miyazaki T, Kuwano H, Clinical Significance and Phenotype of MTA1 Expression in Esophageal Squamous Cell Carcinoma, Anticancer Res 37 (8) (2017) 4147–4155. [DOI] [PubMed] [Google Scholar]
  • [57].Prisco MG, Zannoni GF, De Stefano I, Vellone VG, Tortorella L, Fagotti A, Mereu L, Scambia G, Gallo D, Prognostic role of metastasis tumor antigen 1 in patients with ovarian cancer: a clinical study, Hum Pathol 43 (2) (2012) 282–288. [DOI] [PubMed] [Google Scholar]
  • [58].Kai L, Wang J, Ivanovic M, Chung YT, Laskin WB, Schulze-Hoepfner F, Mirochnik Y, Satcher RL Jr., Levenson AS, Targeting prostate cancer angiogenesis through metastasis-associated protein 1 (MTA1), Prostate 71 (3) (2011) 268–280. [DOI] [PubMed] [Google Scholar]
  • [59].Dias SJ, Zhou X, Ivanovic M, Gailey MP, Dhar S, Zhang L, He Z, Penman AD, Vijayakumar S, Levenson AS, Nuclear MTA1 overexpression is associated with aggressive prostate cancer, recurrence and metastasis in African Americans, Sci Rep 3 (2013) 2331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Kumar R, Wang RA, Mazumdar A, Talukder AH, Mandal M, Yang Z, Bagheri-Yarmand R, Sahin A, Hortobagyi G, Adam L, Barnes CJ, Vadlamudi RK, A naturally occurring MTA1 variant sequesters oestrogen receptor-alpha in the cytoplasm, Nature 418 (6898) (2002) 654–657. [DOI] [PubMed] [Google Scholar]
  • [61].Acconcia F, Kumar R, Signaling regulation of genomic and nongenomic functions of estrogen receptors, Cancer Lett 238 (1) (2006) 1–14. [DOI] [PubMed] [Google Scholar]
  • [62].Dhar S, Kumar A, Zhang L, Rimando AM, Lage JM, Lewin JR, Atfi A, Zhang X, Levenson AS, Dietary pterostilbene is a novel MTA1-targeted chemopreventive and therapeutic agent in prostate cancer, Oncotarget 7 (14) (2016) 18469–18484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Hofer MD, Kuefer R, Varambally S, Li H, Ma J, Shapiro GI, Gschwend JE, Hautmann RE, Sanda MG, Giehl K, Menke A, Chinnaiyan AM, Rubin MA, The role of metastasis-associated protein 1 in prostate cancer progression, Cancer Res 64 (3) (2004) 825–829. [DOI] [PubMed] [Google Scholar]
  • [64].Hofer MD, Tapia C, Browne TJ, Mirlacher M, Sauter G, Rubin MA, Comprehensive analysis of the expression of the metastasis-associated gene 1 in human neoplastic tissue, Arch Pathol Lab Med 130 (7) (2006) 989–996. [DOI] [PubMed] [Google Scholar]
  • [65].Rickman DS, Beltran H, Demichelis F, Rubin MA, Biology and evolution of poorly differentiated neuroendocrine tumors, Nat Med 23 (6) (2017) 1–10. [DOI] [PubMed] [Google Scholar]
  • [66].Beltran H, Prandi D, Mosquera JM, Benelli M, Puca L, Cyrta J, Marotz C, Giannopoulou E, Chakravarthi BV, Varambally S, Tomlins SA, Nanus DM, Tagawa ST, Van Allen EM, Elemento O, Sboner A, Garraway LA, Rubin MA, Demichelis F, Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer, Nat Med 22 (3) (2016) 298–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Kumar A, Dhar S, Campanelli G, Butt NA, Schallheim JM, Gomez CR, Levenson AS, MTA1 drives malignant progression and bone metastasis in prostate cancer, Mol Oncol 12 (9) (2018) 1596–1607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Miyake H, Hara I, Eto H, Serum level of cathepsin B and its density in men with prostate cancer as novel markers of disease progression, Anticancer Res 24 (4) (2004) 2573–2577. [PubMed] [Google Scholar]
  • [69].Podgorski I, Linebaugh BE, Sameni M, Jedeszko C, Bhagat S, Cher ML, Sloane BF, Bone microenvironment modulates expression and activity of cathepsin B in prostate cancer, Neoplasia 7 (3) (2005) 207–223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Ma K, Fan Y, Dong X, Dong D, Guo Y, Wei X, Ning J, Geng Q, Wang C, Hu Y, Li M, Niu W, Li E, Wu Y, MTA1 promotes epithelial to mesenchymal transition and metastasis in non-small-cell lung cancer, Oncotarget 8 (24) (2017) 38825–38840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Cao JM, Li GZ, Han M, Xu HL, Huang KM, MiR-30c-5p suppresses migration, invasion and epithelial to mesenchymal transition of gastric cancer via targeting MTA1, Biomed Pharmacother 93 (2017) 554–560. [DOI] [PubMed] [Google Scholar]
  • [72].Tuncay Cagatay S, Cimen I, Savas B, Banerjee S, MTA-1 expression is associated with metastasis and epithelial to mesenchymal transition in colorectal cancer cells, Tumour Biol 34 (2) (2013) 1189–1204. [DOI] [PubMed] [Google Scholar]
  • [73].Xu C, Hua F, Chen Y, Huang H, Ye W, Yu Y, Shen Z, MTA1 promotes metastasis of MPM via suppression of E-cadherin, J Exp Clin Cancer Res 34 (2015) 151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Wang H, Fan L, Wei J, Weng Y, Zhou L, Shi Y, Zhou W, Ma D, Wang C, Akt mediates metastasis-associated gene 1 (MTA1) regulating the expression of E-cadherin and promoting the invasiveness of prostate cancer cells, PLoS One 7 (12) (2012) e46888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Fan L, Wang H, Xia X, Rao Y, Ma X, Ma D, Wu P, Chen G, Loss of E-cadherin promotes prostate cancer metastasis via upregulation of metastasis-associated gene 1 expression, Oncol Lett 4 (6) (2012) 1225–1233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Dhar S, Kumar A, Gomez CR, Akhtar I, Hancock JC, Lage JM, Pound CR, Levenson AS, MTA1-activated Epi-microRNA-22 regulates E-cadherin and prostate cancer invasiveness, FEBS Lett 591 (6) (2017) 924–933. [DOI] [PubMed] [Google Scholar]
  • [77].Du B, Yang ZY, Zhong XY, Fang M, Yan YR, Qi GL, Pan YL, Zhou XL, Metastasis-associated protein 1 induces VEGF-C and facilitates lymphangiogenesis in colorectal cancer, World J Gastroenterol 17 (9) (2011) 1219–1226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Deng X, Du L, Wang C, Yang Y, Li J, Liu H, Zhang J, Wang L, Zhang X, Li W, Zhang X, Wang S, Dong Z, Close association of metastasis-associated protein 1 overexpression with increased angiogenesis and poor survival in patients with histologically node-negative gastric cancer, World J Surg 37 (4) (2013) 792–798. [DOI] [PubMed] [Google Scholar]
  • [79].Wang T, Li W, Huang H, Wang C, Metastasis-Associated 1 (MTA1) Gene Expression Promotes Angiogenesis in Mouse Xenografts from Human Non-Small Cell Lung Cancer (NSCLC) Cells, Med Sci Monit 25 (2019) 484–491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Qin JH, Ke ZY, Zhou Q, Wang L, Liang Y, Wang YM, Yang T, Gao X, Ye J, Kumar R, Wang RA, Metastasis-Associated Protein 1 Deficiency Results in Compromised Pulmonary Alveolar Capillary Angiogenesis in Mice, Med Sci Monit 23 (2017) 3932–3941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Xue T, Feng W, Yu H, Zhu M, Fei M, Bao Y, Wang X, Ma W, Lv G, Guan J, Chen S, Metastasis-Associated Protein 1 Is Involved in Angiogenesis after Transarterial Chemoembolization Treatment, Biomed Res Int 2017 (2017) 6757898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Nicholson B, Schaefer G, Theodorescu D, Angiogenesis in prostate cancer: biology and therapeutic opportunities, Cancer Metastasis Rev 20 (3–4) (2001) 297–319. [DOI] [PubMed] [Google Scholar]
  • [83].Butt NA, Kumar A, Dhar S, Rimando AM, Akhtar I, Hancock JC, Lage JM, Pound CR, Lewin JR, Gomez CR, Levenson AS, Targeting MTA1/HIF-1alpha signaling by pterostilbene in combination with histone deacetylase inhibitor attenuates prostate cancer progression, Cancer Med 6 (11) (2017) 2673–2685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Li K, Dias SJ, Rimando AM, Dhar S, Mizuno CS, Penman AD, Lewin JR, Levenson AS, Pterostilbene acts through metastasis-associated protein 1 to inhibit tumor growth, progression and metastasis in prostate cancer, PLoS One 8 (3) (2013) e57542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Bostwick DG, Cheng L, Precursors of prostate cancer, Histopathology 60 (1) (2012) 4–27. [DOI] [PubMed] [Google Scholar]
  • [86].Pakala SB, Reddy SD, Bui-Nguyen TM, Rangparia SS, Bommana A, Kumar R, MTA1 coregulator regulates LPS response via MyD88-dependent signaling, J Biol Chem 285 (43) (2010) 32787–32792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Ghanta KS, Pakala SB, Reddy SD, Li DQ, Nair SS, Kumar R, MTA1 coregulation of transglutaminase 2 expression and function during inflammatory response, J Biol Chem 286 (9) (2011) 7132–7138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Bui-Nguyen TM, Pakala SB, Sirigiri RD, Xia W, Hung MC, Sarin SK, Kumar V, Slagle BL, Kumar R, NF-kappaB signaling mediates the induction of MTA1 by hepatitis B virus transactivator protein HBx, Oncogene 29 (8) (2010) 1179–1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Salot S, Gude R, MTA1-mediated transcriptional repression of SMAD7 in breast cancer cell lines, Eur J Cancer 49 (2) (2013) 492–499. [DOI] [PubMed] [Google Scholar]
  • [90].Zhang XY, DeSalle LM, Patel JH, Capobianco AJ, Yu D, Thomas-Tikhonenko A, McMahon SB, Metastasis-associated protein 1 (MTA1) is an essential downstream effector of the c-MYC oncoprotein, Proc Natl Acad Sci U S A 102 (39) (2005) 13968–13973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Powell IJ, Meyskens FL Jr., African American men and hereditary/familial prostate cancer: Intermediate-risk populations for chemoprevention trials, Urology 57 (4 Suppl 1) (2001) 178–181. [DOI] [PubMed] [Google Scholar]
  • [92].Schoonen WM, Salinas CA, Kiemeney LA, Stanford JL, Alcohol consumption and risk of prostate cancer in middle-aged men, Int J Cancer 113 (1) (2005) 133–140. [DOI] [PubMed] [Google Scholar]
  • [93].Jang YG, Go RE, Hwang KA, Choi KC, Resveratrol inhibits DHT-induced progression of prostate cancer cell line through interfering with the AR and CXCR4 pathway, J Steroid Biochem Mol Biol 192 (2019) 105406. [DOI] [PubMed] [Google Scholar]
  • [94].De Amicis F, Chimento A, Montalto FI, Casaburi I, Sirianni R, Pezzi V, Steroid Receptor Signallings as Targets for Resveratrol Actions in Breast and Prostate Cancer, Int J Mol Sci 20 (5) (2019) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Fonseca J, Moradi F, Maddalena LA, Ferreira-Tollstadius B, Selim S, Stuart JA, Resveratrol integrates metabolic and growth effects in PC3 prostate cancer cells-involvement of prolyl hydroxylase and hypoxia inducible factor-1, Oncol Lett 17 (1) (2019) 697–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Cheng TM, Chin YT, Ho Y, Chen YR, Yang YN, Yang YC, Shih YJ, Lin TI, Lin HY, Davis PJ, Resveratrol induces sumoylated COX-2-dependent anti-proliferation in human prostate cancer LNCaP cells, Food Chem Toxicol 112 (2018) 67–75. [DOI] [PubMed] [Google Scholar]
  • [97].Al Aameri RFH, Sheth S, Alanisi EMA, Borse V, Mukherjea D, Rybak LP, Ramkumar V, Tonic suppression of PCAT29 by the IL-6 signaling pathway in prostate cancer: Reversal by resveratrol, PLoS One 12 (5) (2017) e0177198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Kumar S, Eroglu E, Stokes JA 3rd, Scissum-Gunn K, Saldanha SN, Singh UP, Manne U, Ponnazhagan S, Mishra MK, Resveratrol induces mitochondria-mediated, caspase-independent apoptosis in murine prostate cancer cells, Oncotarget 8 (13) (2017) 20895–20908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Taniguchi T, Iizumi Y, Watanabe M, Masuda M, Morita M, Aono Y, Toriyama S, Oishi M, Goi W, Sakai T, Resveratrol directly targets DDX5 resulting in suppression of the mTORC1 pathway in prostate cancer, Cell Death Dis 7 (2016) e2211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Selvaraj S, Sun Y, Sukumaran P, Singh BB, Resveratrol activates autophagic cell death in prostate cancer cells via downregulation of STIM1 and the mTOR pathway, Mol Carcinog 55 (5) (2016) 818–831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Chakraborty A, Gupta N, Ghosh K, Roy P, In vitro evaluation of the cytotoxic, anti-proliferative and anti-oxidant properties of pterostilbene isolated from Pterocarpus marsupium, Toxicol In Vitro 24 (4) (2010) 1215–1228. [DOI] [PubMed] [Google Scholar]
  • [102].Wang TT, Schoene NW, Kim YS, Mizuno CS, Rimando AM, Differential effects of resveratrol and its naturally occurring methylether analogs on cell cycle and apoptosis in human androgen-responsive LNCaP cancer cells, Mol Nutr Food Res 54 (3) (2010) 335–344. [DOI] [PubMed] [Google Scholar]
  • [103].Nikhil K, Sharan S, Chakraborty A, Roy P, Pterostilbene-isothiocyanate conjugate suppresses growth of prostate cancer cells irrespective of androgen receptor status, PLoS One 9 (4) (2014) e93335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Lin VC, Tsai YC, Lin JN, Fan LL, Pan MH, Ho CT, Wu JY, Way TD, Activation of AMPK by pterostilbene suppresses lipogenesis and cell-cycle progression in p53 positive and negative human prostate cancer cells, J Agric Food Chem 60 (25) (2012) 6399–6407. [DOI] [PubMed] [Google Scholar]
  • [105].Kumar A, Dholakia K, Sikorska G, Martinez LA, Levenson AS, MTA1-Dependent Anticancer Activity of Gnetin C in Prostate Cancer, Nutrients 11 (9) (2019) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Narayanan NK, Kunimasa K, Yamori Y, Mori M, Mori H, Nakamura K, Miller G, Manne U, Tiwari AK, 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 4 (11) (2015) 1767–1780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Kumar A, Dhar S, Rimando AM, Lage JM, Lewin JR, Zhang X, Levenson AS, Epigenetic potential of resveratrol and analogs in preclinical models of prostate cancer, Ann N Y Acad Sci 1348 (1) (2015) 1–9. [DOI] [PubMed] [Google Scholar]
  • [108].Luo J, Su F, Chen D, Shiloh A, Gu W, Deacetylation of p53 modulates its effect on cell growth and apoptosis, Nature 408 (6810) (2000) 377–381. [DOI] [PubMed] [Google Scholar]
  • [109].Sankaran D, Pakala SB, Nair VS, Sirigiri DN, Cyanam D, Ha NH, Li DQ, Santhoshkumar TR, Pillai MR, Kumar R, Mechanism of MTA1 protein overexpression-linked invasion: MTA1 regulation of hyaluronan-mediated motility receptor (HMMR) expression and function, J Biol Chem 287 (8) (2012) 5483–5491. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [110].Molli PR, Singh RR, Lee SW, Kumar R, MTA1-mediated transcriptional repression of BRCA1 tumor suppressor gene, Oncogene 27 (14) (2008) 1971–1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Mazumdar A, Wang RA, Mishra SK, Adam L, Bagheri-Yarmand R, Mandal M, Vadlamudi RK, Kumar R, Transcriptional repression of oestrogen receptor by metastasis-associated protein 1 corepressor, Nat Cell Biol 3 (1) (2001) 30–37. [DOI] [PubMed] [Google Scholar]
  • [112].Pakala SB, Bui-Nguyen TM, Reddy SD, Li DQ, Peng S, Rayala SK, Behringer RR, Kumar R, Regulation of NF-kappaB circuitry by a component of the nucleosome remodeling and deacetylase complex controls inflammatory response homeostasis, J Biol Chem 285 (31) (2010) 23590–23597. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [113].Pakala SB, Singh K, Reddy SD, Ohshiro K, Li DQ, Mishra L, Kumar R, TGF-beta1 signaling targets metastasis-associated protein 1, a new effector in epithelial cells, Oncogene 30 (19) (2011) 2230–2241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Reddy SD, Pakala SB, Molli PR, Sahni N, Karanam NK, Mudvari P, Kumar R, Metastasis-associated protein 1/histone deacetylase 4-nucleosome remodeling and deacetylase complex regulates phosphatase and tensin homolog gene expression and function, J Biol Chem 287 (33) (2012) 27843–27850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Balasenthil S, Gururaj AE, Talukder AH, Bagheri-Yarmand R, Arrington T, Haas BJ, Braisted JC, Kim I, Lee NH, Kumar R, Identification of Pax5 as a target of MTA1 in B-cell lymphomas, Cancer Res 67 (15) (2007) 7132–7138. [DOI] [PubMed] [Google Scholar]
  • [116].Talukder AH, Mishra SK, Mandal M, Balasenthil S, Mehta S, Sahin AA, Barnes CJ, Kumar R, MTA1 interacts with MAT1, a cyclin-dependent kinase-activating kinase complex ring finger factor, and regulates estrogen receptor transactivation functions, J Biol Chem 278 (13) (2003) 11676–11685. [DOI] [PubMed] [Google Scholar]
  • [117].Pakala SB, Rayala SK, Wang RA, Ohshiro K, Mudvari P, Reddy SD, Zheng Y, Pires R, Casimiro S, Pillai MR, Costa L, Kumar R, MTA1 promotes STAT3 transcription and pulmonary metastasis in breast cancer, Cancer Res 73 (12) (2013) 3761–3770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Ambros V, microRNAs: tiny regulators with great potential, Cell 107 (7) (2001) 823–826. [DOI] [PubMed] [Google Scholar]
  • [119].Valeri N, Vannini I, Fanini F, Calore F, Adair B, Fabbri M, Epigenetics, miRNAs, and human cancer: a new chapter in human gene regulation, Mamm Genome 20 (9–10) (2009) 573580. [DOI] [PubMed] [Google Scholar]
  • [120].Ventura A, Jacks T, MicroRNAs and cancer: short RNAs go a long way, Cell 136 (4) (2009) 586–591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Schaefer A, Jung M, Mollenkopf HJ, Wagner I, Stephan C, Jentzmik F, Miller K, Lein M, Kristiansen G, Jung K, Diagnostic and prognostic implications of microRNA profiling in prostate carcinoma, Int J Cancer 126 (5) (2010) 1166–1176. [DOI] [PubMed] [Google Scholar]
  • [122].Spahn M, Kneitz S, Scholz CJ, Stenger N, Rudiger T, Strobel P, Riedmiller H, Kneitz B, Expression of microRNA-221 is progressively reduced in aggressive prostate cancer and metastasis and predicts clinical recurrence, Int J Cancer 127 (2) (2010) 394–403. [DOI] [PubMed] [Google Scholar]
  • [123].Clape C, Fritz V, Henriquet C, Apparailly F, Fernandez PL, Iborra F, Avances C, Villalba M, Culine S, Fajas L, miR-143 interferes with ERK5 signaling, and abrogates prostate cancer progression in mice, PLoS One 4 (10) (2009) e7542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Sapre N, Selth LA, Circulating MicroRNAs as Biomarkers of Prostate Cancer: The State of Play, Prostate Cancer 2013 (2013) 539680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Brase JC, Johannes M, Schlomm T, Falth M, Haese A, Steuber T, Beissbarth T, Kuner R, Sultmann H, Circulating miRNAs are correlated with tumor progression in prostate cancer, Int J Cancer 128 (3) (2011) 608–616. [DOI] [PubMed] [Google Scholar]
  • [126].Dhar S, Hicks C, Levenson AS, Resveratrol and prostate cancer: promising role for microRNAs, Mol Nutr Food Res 55 (8) (2011) 1219–1229. [DOI] [PubMed] [Google Scholar]
  • [127].Dhar S, Kumar A, Rimando AM, Zhang X, Levenson AS, Resveratrol and pterostilbene epigenetically restore PTEN expression by targeting oncomiRs of the miR-17 family in prostate cancer, Oncotarget 6 (29) (2015) 27214–27226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Kumar A, Rimando AM, Levenson AS, Resveratrol and pterostilbene as a microRNA-mediated chemopreventive and therapeutic strategy in prostate cancer, Ann N Y Acad Sci 1403 (1) (2017) 15–26. [DOI] [PubMed] [Google Scholar]
  • [129].Mulrane L, McGee SF, Gallagher WM, O’Connor DP, miRNA dysregulation in breast cancer, Cancer Res 73 (22) (2013) 6554–6562. [DOI] [PubMed] [Google Scholar]
  • [130].Scott GK, Mattie MD, Berger CE, Benz SC, Benz CC, Rapid alteration of microRNA levels by histone deacetylase inhibition, Cancer Res 66 (3) (2006) 1277–1281. [DOI] [PubMed] [Google Scholar]
  • [131].Kim W, Benhamed M, Servet C, Latrasse D, Zhang W, Delarue M, Zhou DX, Histone acetyltransferase GCN5 interferes with the miRNA pathway in Arabidopsis, Cell Res 19 (7) (2009) 899–909. [DOI] [PubMed] [Google Scholar]
  • [132].Reddy SD, Pakala SB, Ohshiro K, Rayala SK, Kumar R, MicroRNA-661, a c/EBPalpha target, inhibits metastatic tumor antigen 1 and regulates its functions, Cancer Res 69 (14) (2009) 5639–5642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Avtanski DB, Nagalingam A, Kuppusamy P, Bonner MY, Arbiser JL, Saxena NK, Sharma D, Honokiol abrogates leptin-induced tumor progression by inhibiting Wnt1-MTA1-beta-catenin signaling axis in a microRNA-34a dependent manner, Oncotarget 6 (18) (2015) 16396–16410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Fan C, Lin Y, Mao Y, Huang Z, Liu AY, Ma H, Yu D, Maitikabili A, Xiao H, Zhang C, Liu F, Luo Q, Ouyang G, MicroRNA-543 suppresses colorectal cancer growth and metastasis by targeting KRAS, MTA1 and HMGA2, Oncotarget 7 (16) (2016) 21825–21839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Ferrer P, Asensi M, Segarra R, Ortega A, Benlloch M, Obrador E, Varea MT, Asensio G, Jorda L, Estrela JM, Association between pterostilbene and quercetin inhibits metastatic activity of B16 melanoma, Neoplasia 7 (1) (2005) 37–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Remsberg CM, Yanez JA, Ohgami Y, Vega-Villa KR, Rimando AM, Davies NM, Pharmacometrics of pterostilbene: preclinical pharmacokinetics and metabolism, anticancer, antiinflammatory, antioxidant and analgesic activity, Phytother Res 22 (2) (2008) 169–179. [DOI] [PubMed] [Google Scholar]
  • [137].Kapetanovic IM, Muzzio M, Huang Z, Thompson TN, McCormick DL, Pharmacokinetics, oral bioavailability, and metabolic profile of resveratrol and its dimethylether analog, pterostilbene, in rats, Cancer Chemother Pharmacol 68 (3) (2011) 593–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].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 62 (8) (2014) 1999–2007. [DOI] [PubMed] [Google Scholar]
  • [139].Nakagami Y, Suzuki S, Espinoza JL, Vu Quang L, Enomoto M, Takasugi S, Nakamura A, Nakayama T, Tani H, Hanamura I, Takami A, Immunomodulatory and Metabolic Changes after Gnetin-C Supplementation in Humans, Nutrients 11 (6) (2019) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Lin HS, Yue BD, Ho PC, Determination of pterostilbene in rat plasma by a simple HPLC-UV method and its application in pre-clinical pharmacokinetic study, Biomed Chromatogr 23 (12) (2009) 1308–1315. [DOI] [PubMed] [Google Scholar]
  • [141].Riche DM, McEwen CL, Riche KD, Sherman JJ, Wofford MR, Deschamp D, Griswold M, Analysis of safety from a human clinical trial with pterostilbene, J Toxicol 2013 (2013) 463595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Brown VA, Patel KR, Viskaduraki M, Crowell JA, Perloff M, Booth TD, Vasilinin G, Sen A, Schinas AM, Piccirilli G, Brown K, Steward WP, Gescher AJ, Brenner DE, Repeat dose study of the cancer chemopreventive agent resveratrol in healthy volunteers: safety, pharmacokinetics, and effect on the insulin-like growth factor axis, Cancer Res 70 (22) (2010) 9003–9011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Kjaer TN, Ornstrup MJ, Poulsen MM, Jorgensen JO, Hougaard DM, Cohen AS, Neghabat S, Richelsen B, Pedersen SB, Resveratrol reduces the levels of circulating androgen precursors but has no effect on, testosterone, dihydrotestosterone, PSA levels or prostate volume. A 4-month randomised trial in middle-aged men, Prostate 75 (12) (2015) 1255–1263. [DOI] [PubMed] [Google Scholar]
  • [144].Paller CJ, Rudek MA, Zhou XC, Wagner WD, Hudson TS, Anders N, Hammers HJ, Dowling D, King S, Antonarakis ES, Drake CG, Eisenberger MA, Denmeade SR, Rosner GL, Carducci MA, A phase I study of muscadine grape skin extract in men with biochemically recurrent prostate cancer: Safety, tolerability, and dose determination, Prostate 75 (14) (2015) 1518–1525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].van Die MD, Williams SG, Emery J, Bone KM, Taylor JM, Lusk E, Pirotta MV, A Placebo-Controlled Double-Blinded Randomized Pilot Study of Combination Phytotherapy in Biochemically Recurrent Prostate Cancer, Prostate 77 (7) (2017) 765–775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Von Low EC, Perabo FG, Siener R, Muller SC, Review. Facts and fiction of phytotherapy for prostate cancer: a critical assessment of preclinical and clinical data, In Vivo 21 (2) (2007) 189–204. [PubMed] [Google Scholar]

RESOURCES