ArticlePDF Available

Differential methylation and acetylation as epigenetic basis of Resveratrol’s anticancer activity

Authors:

Abstract and Figures

Numerous studies support the potent anticancer activity of resveratrol and its regulation of key oncogenic signaling pathways. Additionally, the activation of sirtuin 1, a deacetylase, by resveratrol has been known for many years, making resveratrol perhaps one of the earliest nutraceuticals with associated epigenetic activity. Such epigenetic regulation by resveratrol, and the mechanism thereof, has attracted much attention in the past decade. Focusing on methylation and acetylation, the two classical epigenetic regulations, we showcase the potential of resveratrol as an effective anticancer agent by virtue of its ability to induce differential epigenetic changes. We discuss the de-repression of tumor suppressors such as BRCA-1, nuclear factor erythroid 2-related factor 2 (NRF2) and Ras Associated Domain family-1α (RASSF-1α) by methylation, PAX1 by acetylation and the phosphatase and tensin homologue (PTEN) by both methylation and acetylation, in addition to the epigenetic regulation of oncogenic NF-κB and STAT3 signaling by resveratrol. Further, we evaluate the literature supporting the potentiation of HDAC inhibitors and the inhibition of DNMTs by resveratrol in different human cancers. This discussion underlines a robust epigenetic activity of resveratrol that warrants further evaluation, particularly in clinical settings.
Content may be subject to copyright.
medicines
Review
Differential Methylation and Acetylation as the
Epigenetic Basis of Resveratrol’s Anticancer Activity
Mohd Farhan 1, Mohammad Fahad Ullah 2, Mohd Faisal 3, Ammad Ahmad Farooqi 4,
Uteuliyev Yerzhan Sabitaliyevich 5, Bernhard Biersack 6and Aamir Ahmad 7,*
1College of Basic Sciences, King Faisal University, Hofuf 400-Al Ahsa-31982, Saudi Arabia;
mfarhan@kfu.edu.sa
2Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk,
P.O. Box 741, Tabuk 71491, Saudi Arabia; m.ullah@ut.edu.sa
3Department of Psychiatry, University Hospital Limerick, Limerick V94 T9PX, Ireland; mohd.faisal@hse.ie
4Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 44000, Pakistan;
ammadfarooqi@rlmclahore.com
5Department of Postgraduate Education and Research, Kazakhstan Medical University KSPH,
Almaty 050004, Kazakhstan; e.uteuliyev@ksph.kz
6Organic Chemistry Laboratory, Department of Chemistry, University of Bayreuth, Universitaetsstrasse 30,
95447 Bayreuth, Germany; bernhard.biersack@yahoo.com
7Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama,
Mobile, AL 36604, USA
*Correspondence: aahmad@health.southalabama.edu
Received: 11 January 2019; Accepted: 11 February 2019; Published: 13 February 2019


Abstract:
Numerous studies support the potent anticancer activity of resveratrol and its regulation
of key oncogenic signaling pathways. Additionally, the activation of sirtuin 1, a deacetylase,
by resveratrol has been known for many years, making resveratrol perhaps one of the earliest
nutraceuticals with associated epigenetic activity. Such epigenetic regulation by resveratrol, and the
mechanism thereof, has attracted much attention in the past decade. Focusing on methylation and
acetylation, the two classical epigenetic regulations, we showcase the potential of resveratrol as an
effective anticancer agent by virtue of its ability to induce differential epigenetic changes. We discuss
the de-repression of tumor suppressors such as BRCA-1, nuclear factor erythroid 2-related factor 2
(NRF2) and Ras Associated Domain family-1
α
(RASSF-1
α
) by methylation, PAX1 by acetylation and
the phosphatase and tensin homologue (PTEN) by both methylation and acetylation, in addition to the
epigenetic regulation of oncogenic NF-
κ
B and STAT3 signaling by resveratrol. Further, we evaluate
the literature supporting the potentiation of HDAC inhibitors and the inhibition of DNMTs by
resveratrol in different human cancers. This discussion underlines a robust epigenetic activity of
resveratrol that warrants further evaluation, particularly in clinical settings.
Keywords: resveratrol; epigenetic; methylation; acetylation
1. Introduction
Resveratrol (3,5,4
0
-trihydroxy-trans-stilbene) (Figure 1) is a naturally occurring polyphenol
found in peanuts and the skin of grapes and berries. It is a phytoalexin produced in response to
injury, ultraviolet radiation or a pathogen attack. The initial interest in resveratrol was because
of its antioxidant properties [
1
], which led to the recognition of its chemopreventive ability [
2
].
Resveratrol was also found to generate reactive oxygen species, leading to effective anticancer
activity through a prooxidant mechanism [
3
,
4
] which also happens to be a hallmark of several other
polyphenols [
5
7
]. It is now believed that resveratrol exhibits both antioxidant and prooxidant
Medicines 2019,6, 24; doi:10.3390/medicines6010024 www.mdpi.com/journal/medicines
Medicines 2019,6, 24 2 of 12
properties [
8
10
], which depend largely on the tumor microenvironment and the presence of transition
metal ions, particularly copper ions. Besides its anticancer properties, resveratrol has also been
investigated to explain the ‘French paradox’ [
11
], a phenomenon of the relatively low incidence of
coronary heart diseases in the French population despite a diet rich in saturated fats, perhaps due to
the consumption of red wine with high resveratrol content.
Medicines 2019, 6, x FOR PEER REVIEW 2 of 12
depend largely on the tumor microenvironment and the presence of transition metal ions,
particularly copper ions. Besides its anticancer properties, resveratrol has also been investigated to
explain the ‘French paradox’ [11], a phenomenon of the relatively low incidence of coronary heart
diseases in the French population despite a diet rich in saturated fats, perhaps due to the consumption
of red wine with high resveratrol content.
Figure 1. Chemical structure of resveratrol.
The realization of the potent anticancer properties of resveratrol was followed by numerous
investigations into the various signaling pathways that it affects, leading to the observed effects [12–
17]. In recent years, the focus of cancer research has shifted to stem cells, epithelial-to-mesenchymal
transition (EMT) and epigenetic regulation, even in the context of lead compounds such as resveratrol
that have a natural origin. Accordingly, a number of studies have documented the ability of
resveratrol to affect stem cell populations [18,19] and EMT [20,21]. This article focuses on epigenetic
regulation by resveratrol, which is increasingly being proposed as contributing to its anticancer
properties.
2. Epigenetic Regulation by Resveratrol
Even though ‘epigenetics’ was originally intended to define heritable changes in phenotypes
that were independent of changes in the DNA sequence, the meaning of this word has now
considerably broadened to describe the alterations in human chromatin that influence DNA-
templated processes [22,23]. Epigenetic changes do not result in any changes in the DNA sequence,
but can still have lasting effects on gene expression. Epigenetic changes involve at least four known
modifications of DNA and sixteen classes of histone modifications [22–24].
A number of studies have recognized the effects of nutraceuticals, including polyphenol
resveratrol, on the epigenetic machinery in humans [25–35]. Resveratrol can modulate epigenetic
patterns by altering the levels of S-adenosylmethionine and S-adenosylhomocysteine or by directing
the enzymes that catalyze DNA methylation and histone modifications [27]. It also activates the
deacetylase sirtuin and regulates oncogenic and tumor suppressor micro-RNAs [36]. Methylation
and acetylation are two classical epigenetic modifications, and these will be the subject of our
discussion in this article as we showcase the epigenetic basis of the anticancer action of resveratrol.
3. Methylation
The DNA methylation of gene promoters has a profound effect on their eventual expression and
function. Differential DNA methylation can lead to a disease condition in healthy phenotypes. This
is particularly true for cancer wherein the differential DNA methylation of the promoters of
oncogenes and tumor suppressor genes helps maintain a delicate balance. The increased (hyper)
methylation of promoter CpG islands causes gene silencing, while the decreased (hypo) methylation
of promoter CpG islands leads to the expression of the gene.
3.1. Breast Cancer
Breast cancer is a widely studied cancer when it comes to epigenetic regulation, particularly
differential methylation, by resveratrol. Studies have been conducted to elucidate the genome-wide
methylation patterns after resveratrol treatment, and there have also been efforts to understand the
Figure 1. Chemical structure of resveratrol.
The realization of the potent anticancer properties of resveratrol was followed by numerous
investigations into the various signaling pathways that it affects, leading to the observed effects [
12
17
].
In recent years, the focus of cancer research has shifted to stem cells, epithelial-to-mesenchymal
transition (EMT) and epigenetic regulation, even in the context of lead compounds such as resveratrol
that have a natural origin. Accordingly, a number of studies have documented the ability of resveratrol
to affect stem cell populations [
18
,
19
] and EMT [
20
,
21
]. This article focuses on epigenetic regulation by
resveratrol, which is increasingly being proposed as contributing to its anticancer properties.
2. Epigenetic Regulation by Resveratrol
Even though ‘epigenetics’ was originally intended to define heritable changes in phenotypes
that were independent of changes in the DNA sequence, the meaning of this word has now
considerably broadened to describe the alterations in human chromatin that influence DNA-templated
processes [22,23]. Epigenetic changes do not result in any changes in the DNA sequence, but can still
have lasting effects on gene expression. Epigenetic changes involve at least four known modifications
of DNA and sixteen classes of histone modifications [2224].
A number of studies have recognized the effects of nutraceuticals, including polyphenol
resveratrol, on the epigenetic machinery in humans [
25
35
]. Resveratrol can modulate epigenetic
patterns by altering the levels of S-adenosylmethionine and S-adenosylhomocysteine or by directing
the enzymes that catalyze DNA methylation and histone modifications [
27
]. It also activates the
deacetylase sirtuin and regulates oncogenic and tumor suppressor micro-RNAs [
36
]. Methylation and
acetylation are two classical epigenetic modifications, and these will be the subject of our discussion in
this article as we showcase the epigenetic basis of the anticancer action of resveratrol.
3. Methylation
The DNA methylation of gene promoters has a profound effect on their eventual expression and
function. Differential DNA methylation can lead to a disease condition in healthy phenotypes. This is
particularly true for cancer wherein the differential DNA methylation of the promoters of oncogenes
and tumor suppressor genes helps maintain a delicate balance. The increased (hyper) methylation
of promoter CpG islands causes gene silencing, while the decreased (hypo) methylation of promoter
CpG islands leads to the expression of the gene.
3.1. Breast Cancer
Breast cancer is a widely studied cancer when it comes to epigenetic regulation, particularly
differential methylation, by resveratrol. Studies have been conducted to elucidate the genome-wide
methylation patterns after resveratrol treatment, and there have also been efforts to understand the
Medicines 2019,6, 24 3 of 12
epigenetic basis of the de-repression of tumor suppressors by resveratrol. The subsection to follow
will focus on this activity of resveratrol.
3.1.1. Genome-Wide Analyses
A study on the genome-wide methylation-modifying effects of resveratrol, which involved
exposing MCF10CA1h and MCF10CA1a cells to resveratrol for 9 days followed by Illumina 450K
array analysis, revealed a profound effect of resveratrol on genome-wide DNA methylation with
approximately 75% differentially methylated genes being hypermethylated [
37
]. This study evaluated
the epigenetic activity of not only resveratrol but also pterostilbene, an analog of resveratrol.
While treatment with resveratrol was done at a dose of 15
µ
M, pterostilbene treatment was at a
7
µ
M dose. The compounds targeted genes that are over-expressed in tumors because of DNA
hypomethylation. Since increased DNA methylation results in the silencing of genes, it makes sense
that the resveratrol-induced hypermethylated genes have predominantly oncogenic functions [
37
],
such as the Notch signaling pathway. However, in another study that also looked at genome-wide
DNA methylation after resveratrol treatment (in this case, 24 h and 48 h treatment), only about
12.5% of CpG loci were found to be differentially methylated by resveratrol [
38
]. This study used
MDA-MB-231 breast cancer cells and the concentration of resveratrol was 100
µ
M. Additionally, this
study predominantly found DNA hypomethylation by resveratrol. It is possible that the differences
in genome-wide DNA methylation (75% vs. 12.5%) in these two studies might be reflective of the
different experimental setups, in particular, the time-durations of the resveratrol treatment as well as
the doses used, with longer treatment leading to substantially more methylation.
3.1.2. Effects on Tumor Suppressors
Tumor suppressor genes are typically inactivated at the onset of tumorigenesis and, therefore,
their reactivation by anticancer therapy is a mechanism through which tumor progression can be
controlled. Several studies have documented the epigenetic reactivation of the tumor suppressor genes
by resveratrol in breast cancer (Figure 2), as discussed below.
Medicines 2019, 6, x FOR PEER REVIEW 3 of 12
epigenetic basis of the de-repression of tumor suppressors by resveratrol. The subsection to follow
will focus on this activity of resveratrol.
3.1.1. Genome-Wide Analyses
A study on the genome-wide methylation-modifying effects of resveratrol, which involved
exposing MCF10CA1h and MCF10CA1a cells to resveratrol for 9 days followed by Illumina 450K
array analysis, revealed a profound effect of resveratrol on genome-wide DNA methylation with
approximately 75% differentially methylated genes being hypermethylated [37]. This study
evaluated the epigenetic activity of not only resveratrol but also pterostilbene, an analog of
resveratrol. While treatment with resveratrol was done at a dose of 15 μM, pterostilbene treatment
was at a 7 μM dose. The compounds targeted genes that are over-expressed in tumors because of
DNA hypomethylation. Since increased DNA methylation results in the silencing of genes, it makes
sense that the resveratrol-induced hypermethylated genes have predominantly oncogenic functions
[37], such as the Notch signaling pathway. However, in another study that also looked at genome-
wide DNA methylation after resveratrol treatment (in this case, 24 h and 48 h treatment), only about
12.5% of CpG loci were found to be differentially methylated by resveratrol [38]. This study used
MDA-MB-231 breast cancer cells and the concentration of resveratrol was 100 μM. Additionally, this
study predominantly found DNA hypomethylation by resveratrol. It is possible that the differences
in genome-wide DNA methylation (75% vs. 12.5%) in these two studies might be reflective of the
different experimental setups, in particular, the time-durations of the resveratrol treatment as well as
the doses used, with longer treatment leading to substantially more methylation.
3.1.2. Effects on Tumor Suppressors
Tumor suppressor genes are typically inactivated at the onset of tumorigenesis and, therefore,
their reactivation by anticancer therapy is a mechanism through which tumor progression can be
controlled. Several studies have documented the epigenetic reactivation of the tumor suppressor
genes by resveratrol in breast cancer (Figure 2), as discussed below.
Figure 2. Activation of tumor suppressor genes by resveratrol through promoter DNA
hypomethylation. The CpG islands in the DNA promoter regions of the tumor suppressor genes are
hypermethylated resulting in their silencing. De-methylation of these CpG islands by resveratrol
results in the activation of transcription and the eventual expression of tumor suppressor genes such
as the phosphatase and tensin homologue (PTEN), BRCA-1 and nuclear factor erythroid 2-related
factor 2 (NRF2).
Phosphatase and tensin homologue (PTEN) is a well-characterized tumor suppressor in breast
cancer [39]. In one of the earlier studies describing an effect of resveratrol on promoter DNA
methylation, it was reported that resveratrol was highly efficient in reducing PTEN promoter DNA
methylation in MCF7 breast cancer cells [40]. Since reduced DNA methylation leads to gene
expression, this action of resveratrol induced the expression of PTEN which could explain its
anticancer effects. Further, it was reported that the epigenetic effects were complex as not only PTEN
was induced but the cell cycle regulator p21 was up-regulated as well, in addition to the down-
Figure 2.
Activation of tumor suppressor genes by resveratrol through promoter DNA hypomethylation.
The CpG islands in the DNA promoter regions of the tumor suppressor genes are hypermethylated
resulting in their silencing. De-methylation of these CpG islands by resveratrol results in the activation of
transcription and the eventual expression of tumor suppressor genes such as the phosphatase and tensin
homologue (PTEN), BRCA-1 and nuclear factor erythroid 2-related factor 2 (NRF2).
Phosphatase and tensin homologue (PTEN) is a well-characterized tumor suppressor in breast
cancer [
39
]. In one of the earlier studies describing an effect of resveratrol on promoter DNA
methylation, it was reported that resveratrol was highly efficient in reducing PTEN promoter DNA
methylation in MCF7 breast cancer cells [
40
]. Since reduced DNA methylation leads to gene expression,
this action of resveratrol induced the expression of PTEN which could explain its anticancer effects.
Further, it was reported that the epigenetic effects were complex as not only PTEN was induced but
Medicines 2019,6, 24 4 of 12
the cell cycle regulator p21 was up-regulated as well, in addition to the down-regulation of DNMT
(DNA methyltransferase). This is interesting because DNA methyltransferases increase methylation.
Thus, it appears that resveratrol is able to reduce DNA methylation possibly through two different
ways—by reducing methylation and by inducing de-methylation.
BRCA-1 (breast cancer type 1) is another tumor suppressor gene. Its expression is known to
be regulated by epigenetic mechanisms. In a study that demonstrated the effects of resveratrol on
BRCA-1 methylation [
41
], it was shown that exposure of breast cancer cells MCF7 to tumor promoter
TCDD (2,3,7,8 tetrachlorodibenzo-p-dioxin) resulted in the hypermethylation of BRCA-1 promoter CpG
island concomitant with an increased association of trimethylated histone H3K9 and DNMTs, namely
DNMT1, DNMT3a and DNMT3b, with BRCA-1 promoter. Resveratrol, at physiologically relevant
doses, was able to repress these TCDD effects. In a follow-up study, the research group evaluated the
effects of TCDD alone, and in combination with resveratrol, on pregnant Sprague–Dawley rats [
42
].
It was found that, similar to
in vitro
observations in MCF7 cells, gestational exposure to TCDD led
to the reduced DNA CpG island methylation of BRCA-1 promoter in the mammary tissues of the
offspring, which was preceded by the occupation of BRCA-1 promoter by DNMT-1. Also, confirming
a possible therapeutic role, resveratrol was found to partially attenuate TCDD effects [42].
Nuclear factor erythroid 2-related factor 2 (NRF2) is also a tumor suppressor that is
epigenetically regulated by resveratrol. In a study that looked at the effects of resveratrol on E2
(17
β
-estradiol)-induced carcinogenesis [
43
], resveratrol alone up-regulated NRF2 in mammary tissues
of rats, and attenuated the repressive effects of E2 on NRF2. E2 suppressed NRF2 through DNA
methylation, an activity that was inhibited by resveratrol, thus providing further evidence in support
of its regulation of promoter DNA methylation in breast cancer.
3.1.3. DNMTs as Mediators of Resveratrol Effects in Breast Cancer
There is much evidence supporting an inhibitory effect of resveratrol against DNMTs [
40
,
41
,
44
46
].
However, most of the evidence comes from
in vitro
studies. To validate these findings, a pilot study was
conducted that enrolled 39 women with increased breast cancer risk [
47
]. The subjects were divided into
three groups—placebo and those receiving 5 or 50 mg resveratrol. Resveratrol was administered twice
daily for twelve weeks and the focus was on the DNA methylation of four cancer-related genes—p16, Ras
Associated Domain family-1
α
(RASSF-1
α
), Adenomatous Polyposis Coli (APC) and Cyclin D2 (CCND2).
An inverse relationship between serum resveratrol levels and RASSF-1
α
methylation was observed, i.e.,
when resveratrol levels increased, the methylation of RASSF-1
α
decreased, thus leading to the expression of
this tumor suppressor gene. In another study by this group [
48
], an
in vivo
effect of resveratrol on DNMT
expression was examined in normal vs. tumor tissues. The model evaluated was ACI rats, an inbred line
derived from the August and Copenhagen strains. An interesting observation was that resveratrol affected
DNMT3b, but not DNMT1. Two different doses of resveratrol were tested and while DNMT3b differed in
normal vs. tumor tissues of rats treated with low resveratrol, a high resveratrol dose resulted in decreased
DNMT3b in tumor tissues with increased DNMT3b in the normal tissues [
48
]. This observation is a little
different from the one performed
in vitro
in immortalized breast cancer epithelial cells, MCF10A [
49
],
where resveratrol, at a non-cytotoxic dose, only induced subtle changes in the DNA methylation of eight
pre-determined genes. However, the involvement of DNMT3b in resveratrol activity was also identified in
a genome-wide DNA methylation study in breast cancer cells [37].
Further confirming the importance of targeting DNMTs in breast cancer patients, the elevated
expression of DNMT transcripts was reported in a study that evaluated breast cancer tissues from
40 breast cancer patients and compared those with tissues from 10 paired normal breast tissues [
50
].
This study further confirmed the down-regulation of DNMT transcripts
in vitro
in breast cancer cells.
3.2. Glioma
Glioblastoma multiforme was the other malignancy where evidence of the effect of
resveratrol on methylation was initially presented. It was shown that resveratrol could sensitize
Medicines 2019,6, 24 5 of 12
resistant glioblastoma T98G cells to temozolomide, inhibiting temozolomide IC-50 with
increased apoptosis [
51
]. Interestingly, temozolomide-resistant cells have increased MGMT
(O(6)-methylguanine-DNA-methyltransferase) activity and the protein expression of MGMT is
an important determinant of temozolomide-resistance [
51
]. These results are suggestive of yet another
DNA methylation-suppressing activity of resveratrol. In a recent report, inhibition of Wnt signaling
has been identified as the mechanism by which resveratrol induces cell death in T98G cells [
52
].
Epigenetic regulation has been identified as one of the bases of perturbed Wnt signaling [
53
] and it is
plausible that Wnt signaling might be another piece in the puzzle.
3.3. Lung Cancer
Lung cancer, the majority of which is non-small cell lung cancer (NSCLC), is the leading cause of
cancer-related deaths in the United States, as well as worldwide. A number of studies have described
the inhibitory effects of resveratrol against various lung cancer models [
54
56
], albeit, a majority of
the studies have focused on NSCLC with little information on the other lung cancer types. In recent
years, efforts have been made towards the personalized management of lung cancer [
57
], with a
focus on epigenetics in such personalized therapy [
58
]. Further, it has recently been demonstrated
that resveratrol can epigenetically regulate the expression of zinc finger protein36 (ZFP36) through
differential DNA methylation [
59
]. Specifically, resveratrol reduced the methylation of ZFP36, resulting
in its up-regulation in A549 NSCLC cells. A role of ZFP36 in human malignancies is increasingly being
realized, making it an attractive target for therapy [
60
]. Thus, its epigenetic targeting by resveratrol
further underlines the anticancer potential of resveratrol, particularly in lung cancer.
4. Acetylation
Even before the realization of the methylation potential of resveratrol, it has been known to
modulate acetylation within the cellular microenvironment. A quarter century back, resveratrol was
reported to be an activator of sirtuin-1 (SIRT1), the NAD-dependent deacetylase, marking its influence
on acetylation, and thereby epigenetic regulation [
61
]. While the activity of resveratrol against Class
III HDACs sirtuins is well characterized, it has been suggested that perhaps resveratrol possesses
a pan-HDAC inhibitory property and can inhibit HDACs representing class I, class II as well as
class IV [
62
]. Recently, resveratrol has been identified as an inhibitor of bromodomains [
63
] and the
bromodomain and extraterminal (BET) family [
64
]. Bromodomains affect epigenetic machinery by
recognizing lysine acetylation on histones, thereby functioning as epigenetic readers. The interactions
of resveratrol with bromodomains open yet another mechanism of epigenetic regulation by resveratrol
which is not yet fully explored. The next few subsections discuss the reported effects of resveratrol on
the acetylation of different genes in various human cancers.
4.1. Breast Cancer
Acetylation and its impact on breast cancer progression is well appreciated. It is because
of this knowledge that HDAC inhibitors still remain an attractive therapeutic strategy against
breast cancers [
65
67
]. It is not only the HDAC inhibitors but also the inhibitors of HATs (histone
acetyltransferases) that are being evaluated [
68
], which provides a good example of how dynamic
the process of acetylation and deacetylation is, and how an imbalance can lead to tumor onset and
progression. In a study [
69
] that is indicative of an intricate connection between methylation and
acetylation, the two classical epigenetic modifications, it was reported that lysine acetylation within the
signal transducer and activator of transcription 3 (STAT3) can impact the interaction of DNMT1 with
STAT3 and is accompanied by the demethylation and, thereby, the re-expression of tumor suppressor
genes. This study used resveratrol as an acetylation inhibitor and the observations in triple negative
breast cancer (TNBC) were further confirmed in melanoma. TNBCs are characterized by the absence
of the estrogen receptor (ER) and progesterone receptor (PR), as well as human epidermal growth
factor receptor 2 (HER2), and it has been reported that the absence of the ER
α
gene in tumor cells is
Medicines 2019,6, 24 6 of 12
often a result of methylation [
70
]. With the observation that STAT3 is acetylated and, therefore, highly
expressed in TNBCs, it was evaluated whether inhibiting STAT3 acetylation could reactivate ER
α
[
69
].
The TNBC cell line MDA-MB-468 was used as the model and it was observed that treatment with
resveratrol significantly reduced STAT3 acetylation as well as ER
α
gene promoter DNA methylation
(Figure 3). This resulted in the increased expression of ER
α
and the sensitization of otherwise resistant
cells to the ER-targeted therapy, tamoxifen. Further, growth of
in vivo
tumors in mice was not
significantly reduced by tamoxifen alone, as expected, but by the combinational treatment comprising
resveratrol and tamoxifen, thus validating the
in vitro
findings [
69
]. As further evidence of the effect
of resveratrol on acetylation in breast cancer, in MCF-7 breast cancer cells, resveratrol has been shown
to induce H3 acetylation [
71
]. Thus, there seems to be evidence suggesting a modulatory effect of
resveratrol on protein as well as histone acetylation.
Medicines 2019, 6, x FOR PEER REVIEW 6 of 12
[69]. The TNBC cell line MDA-MB-468 was used as the model and it was observed that treatment
with resveratrol significantly reduced STAT3 acetylation as well as ERα gene promoter DNA
methylation (Figure 3). This resulted in the increased expression of ERα and the sensitization of
otherwise resistant cells to the ER-targeted therapy, tamoxifen. Further, growth of in vivo tumors in
mice was not significantly reduced by tamoxifen alone, as expected, but by the combinational
treatment comprising resveratrol and tamoxifen, thus validating the in vitro findings [69]. As further
evidence of the effect of resveratrol on acetylation in breast cancer, in MCF-7 breast cancer cells,
resveratrol has been shown to induce H3 acetylation [71]. Thus, there seems to be evidence suggesting
a modulatory effect of resveratrol on protein as well as histone acetylation.
Figure 3. Epigenetic regulation in triple negative breast cancers (TNBCs). TNBCs are characterized by
activated STAT3 signaling, involving acetylated STAT3. ERα signaling in TNBCs is silenced through
promoter DNA hypermethylation which might be related to STAT3 acetylation but the mechanisms
remain unclear (and are therefore shown with a dotted line). Resveratrol is an effective inhibitor of
STAT3 acetylation as well as ERα promoter DNA methylation. Restoration of ER-signaling makes
TNBC cells sensitive to the ER-targeting therapy, tamoxifen.
4.2. Cervical Cancer
In cervical cancer models, paired box gene1 (PAX1) is a tumor suppressor whose expression is
repressed during tumorigenesis by DNA hypermethylation. In a study that evaluated the effect of
nutraceuticals, including resveratrol, on the inhibition of cervical cancer through the reactivation of
PAX1, it was reported that resveratrol was capable of reactivating PAX1 expression in Caski cells
[72]. However, surprisingly, the reactivation of PAX1 was not found to be due to the effect of
resveratrol on the DNA methylation of PAX1 promoter. Rather, it possibly involved the acetylation
modulating ability of resveratrol through its regulation of HDAC activity. Similar to an effect of
resveratrol on histone H3 acetylation in breast cancer cells above, resveratrol has been reported to
induce H3 acetylation in HeLa cervical cancer cells as well [71]. Such an effect of resveratrol on H3
acetylation assumes significance given the proposed role of histone H3 acetylation as a prognostic
marker for cervical cancer patients [73].
4.3. Colon Cancer
NF-κB signaling is known to be important to the progression of colon cancer [74], especially in
resistance to cisplatin [75]. An increase in the protein acetylation of the NF-κB p65 subunit leads to
the activation of the NF-κB pathway and its nuclear accumulation. Therefore, the inhibition of the
acetylation of p65 can potentially be an effective strategy to check the growth of colon cancer as well
as to overcome resistance to cisplatin. In an in vitro study performed in HK2 cells, resveratrol was
found to decrease the protein acetylation of the p65 subunit, thus reversing the cell viability-inducing
activity of cisplatin [76]. Such down-regulation of NF-κB protein acetylation by resveratrol in
colorectal cells was confirmed in another study [77] and this resulted in reduced tumor invasion and
metastasis because of the down regulation of NF-κB-regulated factors, such as MMP-9 and CXCR4.
4.4. Leukemia and Lymphoma
Figure 3.
Epigenetic regulation in triple negative breast cancers (TNBCs). TNBCs are characterized by
activated STAT3 signaling, involving acetylated STAT3. ER
α
signaling in TNBCs is silenced through
promoter DNA hypermethylation which might be related to STAT3 acetylation but the mechanisms
remain unclear (and are therefore shown with a dotted line). Resveratrol is an effective inhibitor of
STAT3 acetylation as well as ER
α
promoter DNA methylation. Restoration of ER-signaling makes
TNBC cells sensitive to the ER-targeting therapy, tamoxifen.
4.2. Cervical Cancer
In cervical cancer models, paired box gene1 (PAX1) is a tumor suppressor whose expression is
repressed during tumorigenesis by DNA hypermethylation. In a study that evaluated the effect of
nutraceuticals, including resveratrol, on the inhibition of cervical cancer through the reactivation of
PAX1, it was reported that resveratrol was capable of reactivating PAX1 expression in Caski cells [
72
].
However, surprisingly, the reactivation of PAX1 was not found to be due to the effect of resveratrol on
the DNA methylation of PAX1 promoter. Rather, it possibly involved the acetylation modulating ability
of resveratrol through its regulation of HDAC activity. Similar to an effect of resveratrol on histone
H3 acetylation in breast cancer cells above, resveratrol has been reported to induce H3 acetylation in HeLa
cervical cancer cells as well [
71
]. Such an effect of resveratrol on H3 acetylation assumes significance given
the proposed role of histone H3 acetylation as a prognostic marker for cervical cancer patients [73].
4.3. Colon Cancer
NF-
κ
B signaling is known to be important to the progression of colon cancer [
74
], especially in
resistance to cisplatin [
75
]. An increase in the protein acetylation of the NF-
κ
B p65 subunit leads to
the activation of the NF-
κ
B pathway and its nuclear accumulation. Therefore, the inhibition of the
acetylation of p65 can potentially be an effective strategy to check the growth of colon cancer as well
as to overcome resistance to cisplatin. In an
in vitro
study performed in HK2 cells, resveratrol was
found to decrease the protein acetylation of the p65 subunit, thus reversing the cell viability-inducing
activity of cisplatin [
76
]. Such down-regulation of NF-
κ
B protein acetylation by resveratrol in colorectal
cells was confirmed in another study [
77
] and this resulted in reduced tumor invasion and metastasis
because of the down regulation of NF-κB-regulated factors, such as MMP-9 and CXCR4.
Medicines 2019,6, 24 7 of 12
4.4. Leukemia and Lymphoma
In leukemia, resveratrol can potentiate the activity of HDAC inhibitors [
78
], while in a Hodgkin
lymphoma represented by L-428 cells, resveratrol can effectively induce apoptosis as well as cell
cycle arrest [
79
]. As the mechanism, it was observed that resveratrol induced the tumor suppressor
p53 through an increase in the p53 K373-acetylation (Table 1). Additionally, resveratrol treatment was
found to induce the lysine acetylation of FOXO3a [
79
]. In leukemia U937 cells, resveratrol potentiates
reactive oxygen species production by retinoic acid, particularly the production of superoxide anions,
primarily through the up-regulation of the gp91-phox gene that is part of the membrane-bound
cytochrome b
558
[
80
]. As a mechanism, it was elucidated that resveratrol promoted acetylation within
the promoter region of the gp91-phox gene, particularly the Lys-9 residues and Lys-14 residues of
histone H3 within the chromatin surrounding the gene promoter.
Table 1. Epigenetic effects of resveratrol on tumor suppressors: mechanisms of their re-activation.
Tumor Suppressor Cancer Type Effect of Resveratrol Reference
BRCA-1 Breast
Reduced promotor DNA methylation
in vitro [36]
Reduced promotor DNA methylation
in vivo [37]
NRF2 Breast Reduced promotor DNA methylation [38]
p53 Lymphoma Induced acetylation [73]
Prostate [75]
PAX1 Cervical Regulation of histone acetylation [66]
PTEN Breast Reduced promoter DNA methylation [35]
Prostate Acetylation and activation [77]
RASSF-1αBreast Reduced DNA methylation [42]
4.5. Prostate Cancer
In prostate cancer, metastasis-associated protein 1 (MTA1) is oncogenic with its expression
correlating with tumor progression. It is itself involved in the transcriptional repression of target
genes through the post-translational modifications of histones as well as non-histones by virtue of it
being a part of the nucleosome remodeling deacetylation corepressor complex, the ‘NuRD complex’.
Resveratrol was shown to down-regulate MTA1, leading to acetylation and the activation of the
tumor suppressor p53 through the destabilization of the corepressor complex [
81
]. The NuRD
complex plays a role in maintaining chromatin conformation, which it achieves through the
deacetylation of histone proteins [
82
]. HDAC1 and HDA2 are components of the NuRD complex,
with the MTA1-HDAC1 subunit responsible for the deacetylation of histones by NuRD. With a
direct regulation of MTA1, and the observation that HDAC1 was decreased in resveratrol-treated
MTA1 immunoprecipitates, it is evident that resveratrol has a profound effect on histone acetylation.
Further, the effects of resveratrol were similar to those of the HDAC inhibitor SAHA, thus underlying
the acetylation-affecting epigenetic activity of resveratrol. The results were further confirmed
in vivo
in a follow-up study [83], and it was shown that the MTA1-mediated tumor progression was, in part,
due to PTEN inactivation and that resveratrol could acetylate and reactivate PTEN [84] (Table 1).
Signaling through the androgen receptor (AR) is important in prostate cancer, even in the
advanced castrate-resistant prostate cancers. In a study that specifically looked at the regulation of AR
signaling by resveratrol, it was observed that treatment with 10
µ
M resveratrol for just 3 h inhibited
the acetylation of AR and affected the binding of AR to the enhancer region of prostate-specific antigen
(PSA) [
85
]. At a slightly longer treatment of 24 h, resveratrol inhibited the nuclear accumulation of AR
as well. Given the important role that AR plays in prostate cancer, such epigenetic regulation of its
activity and the effect on down-stream signaling by resveratrol is an encouraging finding that gives
hope for its possible use as a therapy against prostate cancer.
Medicines 2019,6, 24 8 of 12
5. Conclusions and Perspectives
While various cellular signaling pathways and genes (oncogenes as well as tumor suppressor
genes) are still being evaluated as therapeutic targets of anticancer agents such as resveratrol, in recent
years, attention has also turned to epigenetic regulation. In fact, epigenetic regulation of classical
signaling pathways is increasingly being realized. For example, two of the very well characterized
signaling pathways, NF-
κ
B and STAT3, are epigenetically regulated by resveratrol [
69
,
76
,
77
,
86
].
This represents a fundamental evolution in our understanding with regards to the intricate regulation
of oncogenic pathways. Our discussion on the topic, as presented in this article, detailed many studies
that provided evidence supporting the epigenetic activity of resveratrol. However, resveratrol does
not regulate gene expression only through epigenetic mechanisms. For example, in a study in acute
lymphoblastic leukemia [
87
] that looked at the possible effect of resveratrol on the methylation of MDR1
(multidrug resistance gene 1), no evidence of the differential DNA methylation of MDR1 promoter by
resveratrol was found. While resveratrol had a visible suppressive effect on MDR1, there did not seem
to be any epigenetic perspective. This is a perfect reminder that not all regulation of gene expression
and function by resveratrol has an epigenetic basis. Additionally, regulation through microRNAs
(miRNAs) is within the realm of epigenetic regulation, but we decided to cover just the classical
epigenetic mechanisms with regards to methylation and acetylation so as to keep the discussion
more focused. Finally, the bioavailability of resveratrol still remains a concern, but that should not
deter us from fully elucidating its mechanism of action and its potential as a viable anticancer lead.
Several groups are working hard on improving the bioavailability through novel approaches and
once they achieve success, resveratrol should be ready for further evaluations in clinical settings as an
anticancer agent with multifaceted epigenetic activity.
Funding:
Mohd Farhan is thankful to the Deanship of Scientific Research, King Faisal University, for a research
grant through the Nasher track (186105).
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Miller, N.J.; Rice-Evans, C.A. Antioxidant activity of resveratrol in red wine. Clin. Chem.
1995
,41, 1789.
[PubMed]
2.
Jang, M.; Cai, L.; Udeani, G.O.; Slowing, K.V.; Thomas, C.F.; Beecher, C.W.; Fong, H.H.; Farnsworth, N.R.;
Kinghorn, A.D.; Mehta, R.G.; et al. Cancer chemopreventive activity of resveratrol, a natural product derived
from grapes. Science 1997,275, 218–220. [CrossRef] [PubMed]
3.
Ahmad, A.; Farhan Asad, S.; Singh, S.; Hadi, S.M. DNA breakage by resveratrol and cu(ii): Reaction
mechanism and bacteriophage inactivation. Cancer Lett. 2000,154, 29–37. [CrossRef]
4.
Ahmad, A.; Syed, F.A.; Singh, S.; Hadi, S.M. Prooxidant activity of resveratrol in the presence of copper ions:
Mutagenicity in plasmid DNA. Toxicol. Lett. 2005,159, 1–12. [CrossRef]
5.
Khan, H.Y.; Zubair, H.; Faisal, M.; Ullah, M.F.; Farhan, M.; Sarkar, F.H.; Ahmad, A.; Hadi, S.M.
Plant polyphenol induced cell death in human cancer cells involves mobilization of intracellular copper
ions and reactive oxygen species generation: A mechanism for cancer chemopreventive action. Mol. Nutr.
Food Res. 2014,58, 437–446. [CrossRef] [PubMed]
6.
Khan, H.Y.; Zubair, H.; Ullah, M.F.; Ahmad, A.; Hadi, S.M. A prooxidant mechanism for the anticancer
and chemopreventive properties of plant polyphenols. Curr. Drug Targets
2012
,13, 1738–1749. [CrossRef]
[PubMed]
7.
Farhan, M.; Oves, M.; Chibber, S.; Hadi, S.M.; Ahmad, A. Mobilization of nuclear copper by green tea
polyphenol epicatechin-3-gallate and subsequent prooxidant breakage of cellular DNA: Implications for
cancer chemotherapy. Int. J. Mol. Sci. 2016,18, 34. [CrossRef] [PubMed]
8.
de la Lastra, C.A.; Villegas, I. Resveratrol as an antioxidant and pro-oxidant agent: Mechanisms and clinical
implications. Biochem. Soc. Trans. 2007,35, 1156–1160. [CrossRef] [PubMed]
Medicines 2019,6, 24 9 of 12
9.
Casey, S.C.; Amedei, A.; Aquilano, K.; Azmi, A.S.; Benencia, F.; Bhakta, D.; Bilsland, A.E.; Boosani, C.S.;
Chen, S.; Ciriolo, M.R.; et al. Cancer prevention and therapy through the modulation of the tumor
microenvironment. Semin. Cancer Biol. 2015,35, S199–S223. [CrossRef] [PubMed]
10.
Chen, C.Y.; Kao, C.L.; Liu, C.M. The cancer prevention, anti-inflammatory and anti-oxidation of bioactive
phytochemicals targeting the tlr4 signaling pathway. Int. J. Mol. Sci. 2018,19, 2729. [CrossRef]
11. Kopp, P. Resveratrol, a phytoestrogen found in red wine. A possible explanation for the conundrum of the
‘french paradox’? Eur. J. Endocrinol. 1998,138, 619–620. [CrossRef] [PubMed]
12.
Shukla, Y.; Singh, R. Resveratrol and cellular mechanisms of cancer prevention. Ann. N. Y. Acad. Sci.
2011
,
1215, 1–8. [CrossRef] [PubMed]
13.
Yang, X.; Li, X.; Ren, J. From french paradox to cancer treatment: Anti-cancer activities and mechanisms of
resveratrol. Anti-Cancer Agents Med. Chem. 2014,14, 806–825. [CrossRef]
14.
McCalley, A.E.; Kaja, S.; Payne, A.J.; Koulen, P. Resveratrol and calcium signaling: Molecular mechanisms
and clinical relevance. Molecules 2014,19, 7327–7340. [CrossRef] [PubMed]
15.
Farooqi, A.A.; Wu, S.J.; Chang, Y.T.; Tang, J.Y.; Li, K.T.; Ismail, M.; Liaw, C.C.; Li, R.N.; Chang, H.W.
Activation and inhibition of atm by phytochemicals: Awakening and sleeping the guardian angel naturally.
Arch. Immunol. Ther. Exp. 2015,63, 357–366. [CrossRef] [PubMed]
16.
de Oliveira, M.R.; Nabavi, S.F.; Manayi, A.; Daglia, M.; Hajheydari, Z.; Nabavi, S.M. Resveratrol and
the mitochondria: From triggering the intrinsic apoptotic pathway to inducing mitochondrial biogenesis,
a mechanistic view. Biochimi. Biophys. Acta 2016,1860, 727–745. [CrossRef] [PubMed]
17.
Zhang, L.; Wen, X.; Li, M.; Li, S.; Zhao, H. Targeting cancer stem cells and signaling pathways by resveratrol
and pterostilbene. BioFactors 2018,44, 61–68. [CrossRef]
18.
Sarkar, F.H.; Li, Y.; Wang, Z.; Kong, D. The role of nutraceuticals in the regulation of wnt and hedgehog
signaling in cancer. Cancer Metast. Rev. 2010,29, 383–394. [CrossRef]
19.
McCubrey, J.A.; Lertpiriyapong, K.; Steelman, L.S.; Abrams, S.L.; Yang, L.V.; Murata, R.M.; Rosalen, P.L.;
Scalisi, A.; Neri, L.M.; Cocco, L.; et al. Effects of resveratrol, curcumin, berberine and other nutraceuticals on
aging, cancer development, cancer stem cells and micrornas. Aging 2017,9, 1477–1536. [CrossRef]
20.
Karimi Dermani, F.; Saidijam, M.; Amini, R.; Mahdavinezhad, A.; Heydari, K.; Najafi, R. Resveratrol inhibits
proliferation, invasion, and epithelial-mesenchymal transition by increasing mir-200c expression in
hct-116 colorectal cancer cells. J. Cell. Biochem. 2017,118, 1547–1555. [CrossRef]
21.
Xu, J.; Liu, D.; Niu, H.; Zhu, G.; Xu, Y.; Ye, D.; Li, J.; Zhang, Q. Resveratrol reverses doxorubicin resistance
by inhibiting epithelial-mesenchymal transition (emt) through modulating pten/akt signaling pathway in
gastric cancer. J. Exp. Clin. Cancer Res. 2017,36, 19. [CrossRef] [PubMed]
22.
Dawson, M.A.; Kouzarides, T. Cancer epigenetics: From mechanism to therapy. Cell
2012
,150, 12–27.
[CrossRef] [PubMed]
23.
Ahmad, A.; Li, Y.; Bao, B.; Kong, D.; Sarkar, F.H. Epigenetic regulation of mirna-cancer stem cells nexus by
nutraceuticals. Mol. Nutr. Food Res. 2014,58, 79–86. [CrossRef] [PubMed]
24.
Baylin, S.B.; Jones, P.A. A decade of exploring the cancer epigenome—biological and translational
implications. Nat. Rev. Cancer 2011,11, 726–734. [CrossRef] [PubMed]
25.
Vanden Berghe, W. Epigenetic impact of dietary polyphenols in cancer chemoprevention: Lifelong
remodeling of our epigenomes. Pharmacol. Res. 2012,65, 565–576. [CrossRef] [PubMed]
26.
Hardy, T.M.; Tollefsbol, T.O. Epigenetic diet: Impact on the epigenome and cancer. Epigenomics
2011
,3,
503–518. [CrossRef] [PubMed]
27.
Park, L.K.; Friso, S.; Choi, S.W. Nutritional influences on epigenetics and age-related disease. Proc. Nutr. Soc.
2012,71, 75–83. [CrossRef] [PubMed]
28.
Gerhauser, C. Cancer chemoprevention and nutriepigenetics: State of the art and future challenges.
Top. Curr. Chem. 2013,329, 73–132. [PubMed]
29.
Wang, Y.; Li, Y.; Liu, X.; Cho, W.C. Genetic and epigenetic studies for determining molecular targets of
natural product anticancer agents. Curr. Cancer Drug Targets 2013,13, 506–518. [CrossRef] [PubMed]
30.
Vahid, F.; Zand, H.; Nosrat-Mirshekarlou, E.; Najafi, R.; Hekmatdoost, A. The role dietary of bioactive
compounds on the regulation of histone acetylases and deacetylases: A review. Gene
2015
,562, 8–15.
[CrossRef] [PubMed]
31.
Gao, Y.; Tollefsbol, T.O. Impact of epigenetic dietary components on cancer through histone modifications.
Curr. Med. Chem. 2015,22, 2051–2064. [CrossRef] [PubMed]
Medicines 2019,6, 24 10 of 12
32.
Kala, R.; Tollefsbol, T.O. A novel combinatorial epigenetic therapy using resveratrol and pterostilbene for
restoring estrogen receptor-alpha (eralpha) expression in eralpha-negative breast cancer cells. PLoS ONE
2016,11, e0155057. [CrossRef] [PubMed]
33.
Fernandes, G.F.S.; Silva, G.D.B.; Pavan, A.R.; Chiba, D.E.; Chin, C.M.; Dos Santos, J.L. Epigenetic regulatory
mechanisms induced by resveratrol. Nutrients 2017,9, 1201. [CrossRef] [PubMed]
34.
Lee, P.S.; Chiou, Y.S.; Ho, C.T.; Pan, M.H. Chemoprevention by resveratrol and pterostilbene: Targeting on
epigenetic regulation. BioFactors 2018,44, 26–35. [CrossRef] [PubMed]
35.
Singh, A.K.; Bishayee, A.; Pandey, A.K. Targeting histone deacetylases with natural and synthetic agents:
An emerging anticancer strategy. Nutrients 2018,10, 731. [CrossRef]
36.
Stefanska, B.; Karlic, H.; Varga, F.; Fabianowska-Majewska, K.; Haslberger, A. Epigenetic mechanisms in
anti-cancer actions of bioactive food components–the implications in cancer prevention. Br. J. Pharmacol.
2012,167, 279–297.
37.
Lubecka, K.; Kurzava, L.; Flower, K.; Buvala, H.; Zhang, H.; Teegarden, D.; Camarillo, I.; Suderman, M.;
Kuang, S.; Andrisani, O.; et al. Stilbenoids remodel the DNA methylation patterns in breast cancer cells
and inhibit oncogenic notch signaling through epigenetic regulation of maml2 transcriptional activity.
Carcinogenesis 2016,37, 656–668. [CrossRef]
38.
Medina-Aguilar, R.; Perez-Plasencia, C.; Marchat, L.A.; Gariglio, P.; Garcia Mena, J.; Rodriguez Cuevas, S.;
Ruiz-Garcia, E.; Astudillo-de la Vega, H.; Hernandez Juarez, J.; Flores-Perez, A.; et al. Methylation landscape
of human breast cancer cells in response to dietary compound resveratrol. PLoS ONE
2016
,11, e0157866.
[CrossRef]
39.
Ramaiah, M.J.; Vaishnave, S. Bmi1 and pten are key determinants of breast cancer therapy: A plausible
therapeutic target in breast cancer. Gene 2018,678, 302–311.
40.
Stefanska, B.; Salame, P.; Bednarek, A.; Fabianowska-Majewska, K. Comparative effects of retinoic acid,
vitamin d and resveratrol alone and in combination with adenosine analogues on methylation and expression
of phosphatase and tensin homologue tumour suppressor gene in breast cancer cells. Br. J. Nutr.
2012
,107,
781–790. [CrossRef]
41.
Papoutsis, A.J.; Borg, J.L.; Selmin, O.I.; Romagnolo, D.F. Brca-1 promoter hypermethylation and silencing
induced by the aromatic hydrocarbon receptor-ligand tcdd are prevented by resveratrol in mcf-7 cells.
J. Nutr. Biochem. 2012,23, 1324–1332. [CrossRef] [PubMed]
42.
Papoutsis, A.J.; Selmin, O.I.; Borg, J.L.; Romagnolo, D.F. Gestational exposure to the ahr
agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin induces brca-1 promoter hypermethylation and reduces
brca-1 expression in mammary tissue of rat offspring: Preventive effects of resveratrol. Mol. Carcinog.
2015,54, 261–269. [CrossRef] [PubMed]
43.
Singh, B.; Shoulson, R.; Chatterjee, A.; Ronghe, A.; Bhat, N.K.; Dim, D.C.; Bhat, H.K. Resveratrol inhibits
estrogen-induced breast carcinogenesis through induction of nrf2-mediated protective pathways.
Carcinogenesis 2014,35, 1872–1880. [CrossRef] [PubMed]
44.
Singh, V.; Sharma, P.; Capalash, N. DNA methyltransferase-1 inhibitors as epigenetic therapy for cancer.
Curr. Cancer Drug Targets 2013,13, 379–399. [CrossRef] [PubMed]
45.
Kala, R.; Shah, H.N.; Martin, S.L.; Tollefsbol, T.O. Epigenetic-based combinatorial resveratrol and
pterostilbene alters DNA damage response by affecting sirt1 and dnmt enzyme expression, including
sirt1-dependent gamma-h2ax and telomerase regulation in triple-negative breast cancer. BMC Cancer
2015
,
15, 672. [CrossRef] [PubMed]
46.
Gao, Y.; Tollefsbol, T.O. Combinational proanthocyanidins and resveratrol synergistically inhibit human
breast cancer cells and impact epigenetic(-)mediating machinery. Int. J. Mol. Sci.
2018
,19, 2204. [CrossRef]
[PubMed]
47.
Zhu, W.; Qin, W.; Zhang, K.; Rottinghaus, G.E.; Chen, Y.C.; Kliethermes, B.; Sauter, E.R. Trans-resveratrol
alters mammary promoter hypermethylation in women at increased risk for breast cancer. Nutr. Cancer
2012
,
64, 393–400. [CrossRef]
48.
Qin, W.; Zhang, K.; Clarke, K.; Weiland, T.; Sauter, E.R. Methylation and mirna effects of resveratrol on
mammary tumors vs. Normal tissue. Nutr. Cancer 2014,66, 270–277. [CrossRef]
49.
Beetch, M.; Lubecka, K.; Kristofzski, H.; Suderman, M.; Stefanska, B. Subtle alterations in DNA methylation
patterns in normal cells in response to dietary stilbenoids. Mol. Nutr. Food Res.
2018
,2018, e1800193.
[CrossRef]
Medicines 2019,6, 24 11 of 12
50.
Mirza, S.; Sharma, G.; Parshad, R.; Gupta, S.D.; Pandya, P.; Ralhan, R. Expression of DNA methyltransferases
in breast cancer patients and to analyze the effect of natural compounds on DNA methyltransferases and
associated proteins. J. Breast Cancer 2013,16, 23–31. [CrossRef]
51.
Huang, H.; Lin, H.; Zhang, X.; Li, J. Resveratrol reverses temozolomide resistance by downregulation of
mgmt in t98g glioblastoma cells by the nf-kappab-dependent pathway. Oncol. Rep.
2012
,27, 2050–2056.
[PubMed]
52.
Yang, H.C.; Wang, J.Y.; Bu, X.Y.; Yang, B.; Wang, B.Q.; Hu, S.; Yan, Z.Y.; Gao, Y.S.; Han, S.Y.; Qu, M.Q.
Resveratrol restores sensitivity of glioma cells to temozolamide through inhibiting the activation of wnt
signaling pathway. J. Cell. Physiol. 2018,2018. [CrossRef]
53.
Koval, A.; Katanaev, V.L. Dramatic dysbalancing of the wnt pathway in breast cancers. Sci. Rep.
2018
,8, 7329.
[CrossRef] [PubMed]
54.
Zhu, Y.; He, W.; Gao, X.; Li, B.; Mei, C.; Xu, R.; Chen, H. Resveratrol overcomes gefitinib resistance by
increasing the intracellular gefitinib concentration and triggering apoptosis, autophagy and senescence in
pc9/g nsclc cells. Sci. Rep 2015,5, 17730. [CrossRef] [PubMed]
55.
Li, W.; Ma, X.; Li, N.; Liu, H.; Dong, Q.; Zhang, J.; Yang, C.; Liu, Y.; Liang, Q.; Zhang, S.; et al.
Resveratrol inhibits hexokinases ii mediated glycolysis in non-small cell lung cancer via targeting akt
signaling pathway. Exp. Cell Res. 2016,349, 320–327. [CrossRef]
56.
Wang, J.; Li, J.; Cao, N.; Li, Z.; Han, J.; Li, L. Resveratrol, an activator of sirt1, induces protective autophagy
in non-small-cell lung cancer via inhibiting akt/mtor and activating p38-mapk. OncoTargets Ther.
2018
,11,
7777–7786. [CrossRef] [PubMed]
57.
Ahmad, A.; Gadgeel, S.M. Lung cancer and personalized medicine: Novel therapies and clinical management.
Preface. Adv. Exp. Med. Biol. 2016,890, v–vi.
58. Ahmad, A. Epigenetics in personalized management of lung cancer. Adv Exp Med Biol 2016,890, 111–122.
59.
Fudhaili, A.; Yoon, N.A.; Kang, S.; Ryu, J.; Jeong, J.Y.; Lee, D.H.; Kang, S.S. Resveratrol epigenetically regulates
the expression of zinc finger protein 36 in nonsmall cell lung cancer cell lines. Oncol. Rep. 2019,41, 1377–1386.
60.
Gupta, G.; Bebawy, M.; Pinto, T.J.A.; Chellappan, D.K.; Mishra, A.; Dua, K. Role of the tristetraprolin (zinc
finger protein 36 homolog) gene in cancer. Crit. Rev. Eukaryot. Gene. Expr. 2018,28, 217–221. [CrossRef]
61.
Howitz, K.T.; Bitterman, K.J.; Cohen, H.Y.; Lamming, D.W.; Lavu, S.; Wood, J.G.; Zipkin, R.E.; Chung, P.;
Kisielewski, A.; Zhang, L.L.; et al. Small molecule activators of sirtuins extend saccharomyces cerevisiae
lifespan. Nature 2003,425, 191–196. [CrossRef] [PubMed]
62.
Venturelli, S.; Berger, A.; Bocker, A.; Busch, C.; Weiland, T.; Noor, S.; Leischner, C.; Schleicher, S.; Mayer, M.;
Weiss, T.S.; et al. Resveratrol as a pan-hdac inhibitor alters the acetylation status of histone [corrected]
proteins in human-derived hepatoblastoma cells. PLoS ONE 2013,8, e73097.
63.
Dutra, L.A.; Heidenreich, D.; Silva, G.; Man Chin, C.; Knapp, S.; Santos, J.L.D. Dietary compound resveratrol
is a pan-bet bromodomain inhibitor. Nutrients 2017,9, 1172. [CrossRef] [PubMed]
64.
Sahni, J.M.; Keri, R.A. Targeting bromodomain and extraterminal proteins in breast cancer. Pharmacol. Res.
2018,129, 156–176. [CrossRef] [PubMed]
65.
Garmpis, N.; Damaskos, C.; Garmpi, A.; Kalampokas, E.; Kalampokas, T.; Spartalis, E.; Daskalopoulou, A.;
Valsami, S.; Kontos, M.; Nonni, A.; et al. Histone deacetylases as new therapeutic targets in triple-negative
breast cancer: Progress and promises. Cancer Genom. Proteom. 2017,14, 299–313.
66.
Damaskos, C.; Garmpis, N.; Valsami, S.; Kontos, M.; Spartalis, E.; Kalampokas, T.; Kalampokas, E.;
Athanasiou, A.; Moris, D.; Daskalopoulou, A.; et al. Histone deacetylase inhibitors: An attractive therapeutic
strategy against breast cancer. Anticancer Res. 2017,37, 35–46. [CrossRef]
67.
Zucchetti, B.; Shimada, A.K.; Katz, A.; Curigliano, G. The role of histone deacetylase inhibitors in metastatic
breast cancer. The Breast 2018,43, 130–134. [CrossRef] [PubMed]
68.
Guo, P.; Chen, W.; Li, H.; Li, M.; Li, L. The histone acetylation modifications of breast cancer and their
therapeutic implications. Pathol. Oncol. Res. 2018,24, 807–813. [CrossRef]
69.
Lee, H.; Zhang, P.; Herrmann, A.; Yang, C.; Xin, H.; Wang, Z.; Hoon, D.S.; Forman, S.J.; Jove, R.; Riggs, A.D.;
et al. Acetylated stat3 is crucial for methylation of tumor-suppressor gene promoters and inhibition by
resveratrol results in demethylation. Proc. Natl. Acad. Sci. USA 2012,109, 7765–7769. [CrossRef]
70.
Lapidus, R.G.; Nass, S.J.; Butash, K.A.; Parl, F.F.; Weitzman, S.A.; Graff, J.G.; Herman, J.G.; Davidson, N.E.
Mapping of er gene cpg island methylation-specific polymerase chain reaction. Cancer Res.
1998
,58,
2515–2519.
Medicines 2019,6, 24 12 of 12
71.
Saenglee, S.; Jogloy, S.; Patanothai, A.; Leid, M.; Senawong, T. Cytotoxic effects of peanut phenolics possessing
histone deacetylase inhibitory activity in breast and cervical cancer cell lines. Pharmacol. Rep.
2016
,68,
1102–1110. [PubMed]
72.
Parashar, G.; Capalash, N. Promoter methylation-independent reactivation of pax1 by curcumin and
resveratrol is mediated by uhrf1. Clin. Exp. Med. 2016,16, 471–478. [CrossRef] [PubMed]
73.
Beyer, S.; Zhu, J.; Mayr, D.; Kuhn, C.; Schulze, S.; Hofmann, S.; Dannecker, C.; Jeschke, U.; Kost, B.P.
Histone h3 acetyl k9 and histone h3 tri methyl k4 as prognostic markers for patients with cervical cancer.
Int. J. Mol. Sci. 2017,18, 477. [CrossRef] [PubMed]
74.
Plewka, D.; Plewka, A.; Miskiewicz, A.; Morek, M.; Bogunia, E. Nuclear factor-kappa b as potential
therapeutic target in human colon cancer. J. Cancer Res. Ther. 2018,14, 516–520. [CrossRef]
75.
Pu, J.; Bai, D.; Yang, X.; Lu, X.; Xu, L.; Lu, J. Adrenaline promotes cell proliferation and increases
chemoresistance in colon cancer ht29 cells through induction of mir-155. Biochem. Biophys. Res. Commun.
2012,428, 210–215. [CrossRef] [PubMed]
76.
Jung, Y.J.; Lee, J.E.; Lee, A.S.; Kang, K.P.; Lee, S.; Park, S.K.; Lee, S.Y.; Han, M.K.; Kim, D.H.; Kim, W.
Sirt1 overexpression decreases cisplatin-induced acetylation of nf-kappab p65 subunit and cytotoxicity in
renal proximal tubule cells. Biochem. Biophys. Res. Commun. 2012,419, 206–210. [CrossRef] [PubMed]
77.
Buhrmann, C.; Shayan, P.; Popper, B.; Goel, A.; Shakibaei, M. Sirt1 is required for resveratrol-mediated
chemopreventive effects in colorectal cancer cells. Nutrients 2016,8, 145. [CrossRef]
78.
Yaseen, A.; Chen, S.; Hock, S.; Rosato, R.; Dent, P.; Dai, Y.; Grant, S. Resveratrol sensitizes acute myelogenous
leukemia cells to histone deacetylase inhibitors through reactive oxygen species-mediated activation of the
extrinsic apoptotic pathway. Mol. Pharmacol. 2012,82, 1030–1041. [CrossRef]
79.
Frazzi, R.; Valli, R.; Tamagnini, I.; Casali, B.; Latruffe, N.; Merli, F. Resveratrol-mediated apoptosis of hodgkin
lymphoma cells involves sirt1 inhibition and foxo3a hyperacetylation. Int. J. Cancer
2013
,132, 1013–1021.
[CrossRef]
80.
Kikuchi, H.; Mimuro, H.; Kuribayashi, F. Resveratrol strongly enhances the retinoic acid-induced superoxide
generating activity via up-regulation of gp91-phox gene expression in u937 cells. Biochem. Biophys.
Res. Commun. 2018,495, 1195–1200. [CrossRef]
81.
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. [CrossRef] [PubMed]
82.
Xue, Y.; Wong, J.; Moreno, G.T.; Young, M.K.; Cote, J.; Wang, W. Nurd, a novel complex with both
atp-dependent chromatin-remodeling and histone deacetylase activities. Mol. Cell
1998
,2, 851–861.
[CrossRef]
83.
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. [CrossRef] [PubMed]
84.
Dhar, S.; Kumar, A.; Li, K.; Tzivion, G.; Levenson, A.S. Resveratrol regulates pten/akt pathway through
inhibition of mta1/hdac unit of the nurd complex in prostate cancer. Biochim. Biophys. Acta
2015
,1853,
265–275. [CrossRef] [PubMed]
85.
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. [CrossRef]
86.
Chao, S.C.; Chen, Y.J.; Huang, K.H.; Kuo, K.L.; Yang, T.H.; Huang, K.Y.; Wang, C.C.; Tang, C.H.; Yang, R.S.;
Liu, S.H. Induction of sirtuin-1 signaling by resveratrol induces human chondrosarcoma cell apoptosis and
exhibits antitumor activity. Sci. Rep. 2017,7, 3180. [CrossRef]
87.
Zadi Heydarabad, M.; Nikasa, M.; Vatanmakanian, M.; Azimi, A.; Farshdousti Hagh, M. Regulatory effect of
resveratrol and prednisolone on mdr1 gene expression in acute lymphoblastic leukemia cell line (ccrf-cem):
An epigenetic perspective. J. Cell Biochem. 2018,119, 4890–4896. [CrossRef]
©
2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... The observed protective effects of O. variegata extract may be attributed to its rich phytochemical composition, which includes a diverse array of bioactive compounds including phenolic compounds, flavonoids, and tannins (Dias, Pinto, and Silva 2021;Farhan et al. 2019;Farooqi et al. 2020;Rana et al. 2022;Wahnou, Limami, and Oudghiri 2024;X. Zeng et al. 2023). ...
Article
Skin cancer is the most widespread type of malignant tumor representing a major public health concern. Considering the numerous side effects associated with conventional treatments, phytotherapy may be regarded as a viable medicinal alternative. This study aimed to investigate the therapeutic potential of Orbea variegata (L.) Haw, an ornamental plant, in treating skin cancer using an animal model induced by a combination of ultraviolet (UV) irradiation and sulfuric acid treatment. The hydroethanolic extract of Orbea variegata underwent phytochemical characterization, identifying the presence of reducing sugars, coumarins, alkaloids, flavonoids, tannins, and saponins through qualitative screening. Quantitative analysis demonstrated significant amounts of phenolic compounds (29.435 ± 0.571 mg GAE/g of dry extract), flavonoids (6.711 ± 0.272 mg QE/g of dry extract), and tannins (274.037 ± 11.3 mg CE/g of dry extract). The administration the hydroethanolic extract in two concentrations (1 or 2 g/kg) to male Swiss mice exhibited no marked adverse effects, as evidenced by serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) enzyme activity levels. In addition, the extract significantly reduced skin hyperplasia and inflammation induced by UV/sulfuric acid treatment as noted in tissue analyses and decreased protein expression of nuclear proliferation marker (Ki-67). This improvement was associated with a marked decrease in oxidative stress, as indicated by diminished lipid peroxidation levels, and restoration of the activity of endogenous antioxidant enzyme catalase (CAT) to control levels. Our findings demonstrated the potential of Orbea variegata hydroethanolic extract to be considered as a treatment for skin cancer, exhibiting its apparent safety and efficacy in reducing inflammation and carcinogenesis in a UV/sulfuric acid-induced Swiss mouse model, attributed to its phytochemical content and associated antioxidant activities.
... Due to increasing multidrug resistance in cancer, numerous studies are now using natural products to target NF-κB and induce apoptosis or autophagy, leading to cancer suppression [3,16,[29][30][31][32][33]. Furthermore, many of these phytochemicals, such as curcumin, resveratrol, and lycopene, exert their anti-cancer activity not only by target-ing NF-κB but also by controlling epigenetic modifications, which are key regulators of apoptosis and autophagy [30,[32][33][34][35][36]. ...
Article
Full-text available
Naphthylisoquinoline (NIQ) alkaloids are rising as a promising class of secondary metabolites with pharmaceutical potential. NF-κB has already been recognized as a significant modulator of cancer proliferation and drug resistance. We have previously reported the mechanisms behind the cytotoxic effect of dioncophylline A, an NIQ monomer, in leukemia cells. In the current study, we have investigated the cytotoxic effect of jozimine A2, an NIQ dimer, on leukemia cells in comparison to a second, structurally unsymmetric dimer, michellamine B. To this end, molecular docking was applied to predict the binding affinity of the dimers towards NF-κB, which was then validated through microscale thermophoresis. Next, cytotoxicity assays were performed on CCRF-CEM cells and multidrug-resistant CEM/ADR5000 cells following treatment. Transcriptome analysis uncovered the molecular networks affected by jozimine A2 and identified the cell cycle as one of the major affected processes. Cell death modes were evaluated through flow cytometry, while angiogenesis was measured with the endothelial cell tube formation assay on human umbilical vein endothelial cells (HUVECs). The results indicated that jozimine A2 bound to NF-κB, inhibited its activity and prevented its translocation to the nucleus. In addition, jozimine A2 induced cell death through apoptosis and prevented angiogenesis. Our study describes the cytotoxic effect of jozimine A2 on leukemia cells and explains the interactions with the NF-κB signaling pathway and the anticancer activity.
... For example, other natural compounds, like resveratrol, EGCG, sulforaphane, genistein, and quercetin, have all demonstrated potential in modifying epigenetic processes, thereby influencing cancer cell behavior [138,139]. Resveratrol, found in grapes, red wine, and berries, has been shown to affect various epigenetic processes [141][142][143]. EGCG [134], a major active component of green tea; sulforaphane [144], found in cruciferous vegetables; genistein [145], a soy isoflavone; and quercetin [146], a flavonoid found in many fruits and vegetables, have all shown potential anticancer activity by affecting epigenetic processes via regulation of DNA methylation and histone modification in cancer cells. ...
Article
Full-text available
While significant strides have been made in understanding cancer biology, the enhancement in patient survival is limited, underscoring the urgency for innovative strategies. Epigenetic modifications characterized by hereditary shifts in gene expression without changes to the DNA sequence play a critical role in producing alternative gene isoforms. When these processes go awry, they influence cancer onset, growth, spread, and cancer stemness. In this review, we delve into the epigenetic and isoform nuances of the protein kinase, doublecortin-like kinase 1 (DCLK1). Recognized as a hallmark of tumor stemness, DCLK1 plays a pivotal role in tumorigenesis, and DCLK1 isoforms, shaped by alternative promoter usage and splicing, can reveal potential therapeutic touchpoints. Our discussion centers on recent findings pertaining to the specific functions of DCLK1 isoforms and the prevailing understanding of its epigenetic regulation via its two distinct promoters. It is noteworthy that all DCLK1 isoforms retain their kinase domain, suggesting that their unique functionalities arise from non-kinase mechanisms. Consequently, our research has pivoted to drugs that specifically influence the epigenetic generation of these DCLK1 isoforms. We posit that a combined therapeutic approach, harnessing both the epigenetic regulators of specific DCLK1 isoforms and DCLK1-targeted drugs, may prove more effective than therapies that solely target DCLK1.
... Resveratrol may modulate DNA methylation by altering the levels of S-adenosylmethionine and S-adenosylhomocysteine or by affecting the enzymes that catalyze DNA methylation [77]. ...
Article
Healthy aging can be defined as an extended lifespan and health span. Nutrition has been regarded as an important factor in healthy aging, because nutrients, bioactive food components, and diets have demonstrated beneficial effects on aging hallmarks such as oxidative stress, mitochondrial function, apoptosis and autophagy, genomic stability, and immune function. Nutrition also plays a role in epigenetic regulation of gene expression, and DNA methylation is the most extensively investigated epigenetic phenomenon in aging. Interestingly, age-associated DNA methylation can be modulated by one-carbon metabolism or inhibition of DNA methyltransferases. One-carbon metabolism ultimately controls the balance between the universal methyl donor S-adenosylmethionine and the methyltransferase inhibitor S-adenosylhomocysteine. Water-soluble B-vitamins such as folate, vitamin B6, and vitamin B12 serve as coenzymes for multiple steps in one-carbon metabolism, whereas methionine, choline, betaine, and serine act as methyl donors. Thus, these one-carbon nutrients can modify age-associated DNA methylation and subsequently alter the age-associated physiologic and pathologic processes. We cannot elude aging per se but we may at least change age-associated DNA methylation, which could mitigate age-associated diseases and disorders.
... In addition, resveratrol has been shown to induce the reactivation of silenced tumor suppressor genes, such as RARβ, in cancer cells. Resveratrol has shown promise as a potential therapeutic agent for various types of cancer (Farhan et al., 2019). Resveratrol inhibits the proliferation and induces apoptosis in meningioma cells by upregulating miR-34a-3p (Hu et al., 2019). ...
Article
Full-text available
Meningioma, one of the most common primary central nervous system tumors, are classified into three grades by the World Health Organization (WHO) based on histopathology. The gold-standard treatment, surgical resection, is hampered by issues such as incomplete resection in some cases and a high recurrence rate. Alongside genetic alterations, DNA methylation, plays a crucial role in progression of meningiomas in the occurrence and development of meningiomas. The epigenetic landscape of meningioma is instrumental in refining tumor classification, identifying robust molecular markers, determining prognosis, guiding treatment selection, and innovating new therapeutic strategies. Existing classifications lack comprehensive accuracy, and effective therapies are limited. Methylated DNA markers, exhibiting differential characteristics across varying meningioma grades, serve as invaluable diagnostic tools. Particularly, combinatorial methylated markers offer insights into meningioma pathogenesis, tissue origin, subtype classification, and clinical outcomes. This review integrates current research to highlight some of the most promising DNA and promoter methylation markers employed in meningioma diagnostics. Despite their promise, the development and application of DNA methylation biomarkers for meningioma diagnosis and treatment are still in their infancy, with only a handful of DNA methylation inhibitors currently clinically employed for meningioma treatment. Future studies are essential to validate these markers and ascertain their clinical utility. Combinatorial methylated DNA markers for meningiomas have broad implications for understanding tumor development and progression, signaling a paradigm shift in therapeutic strategies for meningiomas.
... The incidence of skin related diseases has increased with industrialization and pollution because these environmental factors alter epigenetic regulation [56]. Gene expression is controlled by non-coding RNAs, DNA methylation and histone modification [57][58][59]. In AD, there has been evidence of altered genetic and epigenetic expression in immune cells, which contributes to disease pathogenesis [56,60]. ...
Article
Atopic dermatitis (AD), also known as atopic eczema, is a common but also complex chronic, itchy skin condition with underlying inflammation of the skin. This skin ailment is prevalent worldwide and affects people of all ages, particularly children below five years of age. The itching and resulting rashes in AD patients are often the result of inflammatory signals, thus necessitating a closer look at the inflammation-regulating mechanisms for putative relief, care and therapy. Several chemical- as well as genetically-induced animal models have established the importance of targeting pro-inflammatory AD microenvironment. Epigenetic mechanisms are gaining attention towards a better understanding of the onset as well as the progression of inflammation. Several physiological processes with implications in pathophysiology of AD, such as, barrier dysfunction either due to reduced filaggrin / human β-defensins or altered microbiome, reprograming of Fc receptors with resulting overexpression of high affinity IgE receptors, elevated eosinophil numbers or the elevated IL-22 production by CD4 + T cells have underlying epigenetic mechanisms that include differential promoter methylation and/or regulation by non-coding RNAs. Reversing these epigenetic changes has been verified to reduce inflammatory burden through altered secretion of cytokines IL-6, IL-4, IL-13, IL-17, IL-22 etc, with benefit against AD progression in experimental models. A thorough understanding of epigenetic remodeling of inflammation in AD has the potential of opening avenues for novel diagnostic, prognostic and therapeutic options.
Chapter
Epigenetics has gained popularity as a promising area for nutritional intervention. Epigenetic patterns are set during early life, and this critical period is exceptionally responsive to environmental factors. Dietary metabolites and gut microbiota directly or through interactions via epigenetic modifications contribute to the development of various diseases. In this study, relevant literature has been reviewed to understand the connection among nutrition, epigenetics, and gut microbiota, and their impact on health and disease.
Chapter
The role of epigenetics, focusing on changes in gene functions unrelated to DNA sequence, is becoming increasingly evident in cancer initiation, progression, metastasis and treatment. Here, we highlight the role of chromatin, DNA methylation, histone acetylation and histone methylation in gene regulation and their contribution to oncogenesis. The paper examines the potential of targeting epigenetic modifications like DNMTs, HDACs and HMTs as cancer treatments, encompassing their impact on gene expression and various apoptotic pathways as well as their clinical implications. Recently, the use of this knowledge has resulted in various FDA-approved drugs such as 5-azacytidine, suberoylanilide hydroxamic acid and many more. Additionally, it explores how traditional medicines rich in flavonoids contain active compounds such as kaempferol, curcumin and quercetin, which could affect cancer-related epigenetic changes. The need for validation, standardisation and collaboration between traditional healers and scientists is stressed, concluding that merging traditional medicine with scientific research holds the potential for advancing cancer therapies.
Article
Full-text available
Neurodegeneration is a process leading to the progressive loss of structure and functions of neurons. Many neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease have shown many common points at the subcellular level. Neurons are metabolically active cells and need a high amount of energy. Mitochondria are known as the energy synthesis center for cells, involved in the synthesis of adenosine triphosphate by oxidative phosphorylation. Rather than just being an energy synthesis center, it has critical importance for many cellular functions such as calcium homeostasis, cell proliferation, cell growth, and apoptosis. In the process of mitochondrial dysfunction, cellular functions are disrupted and cells enter the apoptotic or necrotic pathway. Resveratrol (trans-3,5,4-trihydoxystilbene), a plant-derived polyphenol found in the seed of grapes, berries, peanuts, and wine, has many biological effects such as inhibition of lipid peroxidation, scavenging of free radicals, changes in eicosanoid synthesis, inhibition of platelet aggregation, anti-inflammatory and anticancer activity, and regulation of lipid metabolism. Through the reviewed literature, the current study investigated the protective role of resveratrol in neurodegenerative diseases. Studies show that resveratrol moderates mitochondrial function, redox status, and cellular dynamics in both in vivo and in vitro experimental models of neurodegeneration. Resveratrol suppresses reactive oxygen species production by reducing the activity of complex III due to its competition effect with coenzyme Q. In the present work, we discussed the protective effects of resveratrol on neurodegeneration, neurodegenerative diseases, and the redox biology of the mitochondria.
Article
The hormone producing hypothalamus, pituitary and gonadal are arranged in hierarchy to form the hypothalamic-pituitary-gonadal axis (HPG axis). The axis is neuroendocrine in nature and releases hormones in response to the inputs from nervous systems. The axis maintains homeostasis and ensures smooth body functions, particularly those related to growth and reproduction. A deregulated HPG axis, such as observed under inflammation and other conditions, is therefore associated with several disorders such as polycystic ovary syndrome, functional hypothalamic amenorrhea etc. Several factors, both genetic as well as environmental, in addition to aging, obesity etc. affect HPG axis with resulting effects on puberty, sexual maturation and reproductive health. More research is now indicative of a role of epigenetics in mediating these HPG-affecting factors. Hypothalamus-secreted gonadotropin-releasing hormone is important for eventual release of sex hormones and it is subjected to several neuronal and epigenetic regulations. Gene promoter methylation as well as histone methylations and acetylations form the backbone of epigenetic regulation of HPG-axis, as the incoming reports suggest. Epigenetic events also mediate several feedback mechanisms within HPG axis and between HPG axis and the central nervous system. In addition, data is emerging for a role of non-coding RNAs, particularly the miRNAs, in regulation and normal functioning of HPG axis. Thus, the epigenetic interactions need better understanding to understand the functioning and regulation of HPG axis.
Article
Full-text available
Background: Resveratrol, a natural polyphenolic phytoalexin, has potent anti-tumor activity. Recently, it was found to induce autophagy in cancer cells. However, the effects of resveratrol on autophagy in non-small-cell lung cancer (NSCLC) cells have not yet been clearly elucidated. Materials and methods: A549 and H1299 cells were treated with different concentrations of resveratrol. Cell growth and apoptosis were measured by CCK-8 assay and flow cytometry, respectively. A549 cells were then treated with 200 μM resveratrol or SRT1720. Cell autophagy was detected by western blot and immunofluorescence. Results: In this study, we found that resveratrol exerted the anti-tumor effect through inhibiting cell proliferation and promoting cell apoptosis in NSCLC cells dose-dependently. Resveratrol has also increased the relative expression of Beclin1 and LC3 II/I while decreased p62 expression, suggesting that resveratrol induced autophagy in NSCLC cells. In addition, resveratrol increased SIRT1 expression and SIRT1 activator SRT1720-induced autophagy of NSCLC cells. SIRT1 knockdown reduced resveratrol-induced autophagy significantly. These results indicated that resveratrol might induce autophagy through upregulating SIRT1 expression. Moreover, inhibiting autophagy by autophagy inhibitor 3-methyladenine or SIRT1 inhibitor nicotinamide significantly suppressed proliferation while promoted apoptosis compared with the resveratrol 200 μM group, suggesting that resveratrol-induced autophagy might act as a protective mechanism to promote NSCLC cell survival and inhibiting autophagy can enhance the anti-tumor effect of resveratrol. Besides that, resveratrol treatment inhibited Akt/mTOR while p38-MAPK was activated in NSCLC cells in a dose-dependent manner. Activating Akt/ mTOR pathway by IGF-1 or inhibiting p-38-MAPK pathway by doramapimod significantly inhibited cell proliferation while increased cell apoptosis of NSCLC cells compared with the resveratrol 200 μM group. Conclusion: Taken together, our findings suggest that resveratrol inhibited proliferation but induced apoptosis and autophagy via inhibiting Akt/mTOR and activating p38-MAPK pathway. Resveratrol-induced autophagy might act as a protective mechanism to promote NSCLC cell survival. Therefore, inhibition of autophagy may enhance the anti-tumor activity of resveratrol in NSCLC.
Article
Full-text available
Toll-like receptors (TLRs) are a well-known family of pattern recognition receptors that play an important role in a host immune system. TLR triggering leads to the induction of pro-inflammatory cytokines and chemokines, driving the activation of both innate and adaptive immunity. Recently, an increasing number studies have shown the link between TLRs and cancer. Among them, the toll-like receptor 4 (TLR4) signaling pathway is associated with inflammatory response and cancer progression. Dietary phytochemicals are potential modulators of immunological status with various pharmacological properties including anti-cancer, anti-oxidant and anti-inflammatory. Curcumin, 6-gingerol, 6-shogaol, 1-dehydro-10-gingerdione, epigallocatechin gallate (EGCG), luteolin, quercetin, resveratrol, caffeic acid phenethyl ester, xanthohumol, genistein, berberine, and sulforaphane can inhibit TLR4 activation. The aim of the present review is to describe the role of the TLR4 signaling pathway between inflammatory response and cancer progression. We further introduce bioactive phytochemicals with potential anti-inflammation and chemoprevention by inhibiting TLR activation.
Article
Full-text available
Breast cancer is the second most common cancer and the second leading cause of death from cancer among women in the United States (US). Cancer prevention and therapy through the use of phytochemicals that have epigenetic properties has gained considerable interest during the past few decades. Such dietary components include, but are not limited to, grape seed proanthocyanidins (GSPs) and resveratrol (Res), both of which are present in red wine. In this study, we report for the first time the synergistic effects of GSPs and Res on inhibiting MDA-MB-231 and MCF-7 human breast cancer cells. Our results of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays and clonogenic assays indicate that treatments with the combinations of GSPs and Res synergistically decreased cell viability and posttreatment cell proliferation in both cell lines. Additional analyses show that treatments with GSPs and Res in combination synergistically induced apoptosis in MDA-MB-231 cells by upregulating Bax expression and down-regulating Bcl-2 expression. DNA methyltransferase (DNMT) activity and histone deacetylase (HDAC) activity were greatly reduced in MDA-MB-231 and MCF-7 cells after treatments with GSPs and Res in combination. Collectively, our findings suggest that GSPs and Res synergistically inhibit human breast cancer cells through inducing apoptosis, as well as modulating DNA methylation and histone modifications.
Article
Full-text available
Cancer initiation and progression are the result of genetic and/or epigenetic alterations. Acetylation-mediated histone/non-histone protein modification plays an important role in the epigenetic regulation of gene expression. Histone modification is controlled by the balance between histone acetyltransferase and (HAT) and histone deacetylase (HDAC) enzymes. Imbalance between the activities of these two enzymes is associated with various forms of cancer. Histone deacetylase inhibitors (HDACi) regulate the activity of HDACs and are being used in cancer treatment either alone or in combination with other chemotherapeutic drugs/radiotherapy. The Food and Drug Administration (FDA) has already approved four compounds, namely vorinostat, romidepsin, belinostat, and panobinostat, as HDACi for the treatment of cancer. Several other HDACi of natural and synthetic origin are under clinical trial for the evaluation of efficiency and side-effects. Natural compounds of plant, fungus, and actinomycetes origin, such as phenolics, polyketides, tetrapeptide, terpenoids, alkaloids, and hydoxamic acid, have been reported to show potential HDAC-inhibitory activity. Several HDACi of natural and dietary origin are butein, protocatechuic aldehyde, kaempferol (grapes, green tea, tomatoes, potatoes, and onions), resveratrol (grapes, red wine, blueberries and peanuts), sinapinic acid (wine and vinegar), diallyl disulfide (garlic), and zerumbone (ginger). HDACi exhibit their antitumor effect by the activation of cell cycle arrest, induction of apoptosis and autophagy, angiogenesis inhibition, increased reactive oxygen species generation causing oxidative stress, and mitotic cell death in cancer cells. This review summarizes the HDACs classification, their aberrant expression in cancerous tissue, structures, sources, and the anticancer mechanisms of HDACi, as well as HDACi that are either FDA-approved or under clinical trials.
Article
Histone deacetylase inhibitors (HDACi) are a relatively new class of drug that plays an important role in the epigenetic and non-epigenetic regulation in cancer, inducing death, apoptosis and cell cycle arrest in cancer cells. Although HDACi are approved only for hematologic malignancies, there are several trials in the breast cancer setting with promising results. In this review, we summarize the latest studies with HDACi in breast cancer from the emerging data in the translational research until its possible applicability in the clinical practice.
Article
Zinc finger protein 36 (ZFP36) is an AU‑rich element protein that binds to 3'‑untranslated regions and promotes the decay of target mRNAs. Downregulation of ZFP36 expression in turn results in stabilization of target mRNAs. A recent study indicated that downregulation of ZFP36 expression in human liver cancer is caused by epigenetic mechanisms. The purpose of the present study was to investigate the potential of resveratrol (Res) to induce ZFP36 expression. Promoter methylation was analyzed using methylation‑sensitive restriction analysis. It was determined that Res treatment increased ZFP36 expression and decreased the mRNA levels of ZFP36 target genes in A549 lung cancer cells. Additionally, Res suppressed the expression of DNA (cytosine‑5)‑methyltransferase 1 and induced demethylation of the ZFP36 promoter. Collectively, the present results demonstrated that Res has anticancer activity through its epigenetic regulation of ZFP36 in non‑small cell lung cancer.
Article
Cancer is a complicated transformational progression that fiercely changes the appearance of cell physiology as well as cells' relations with adjacent tissues. Developing an oncogenic characteristic requires a wide range of modifications in a gene expression at a cellular level. This can be achieved by activation or suppression of the gene regulation pathway in a cell. Tristetraprolin (TTP or ZFP36) associated with the initiation and development of tumors are regulated at the level of mRNA decay, frequently through the activity of AU-rich mRNA-destabilizing elements (AREs) located in their 3'-untranslated regions. TTP is an attractive target for therapeutic use and diagnostic tools due to its characteristic appearance in cancer tissue alone. Thus, the illumination of TTP in diverse types of cancer might deliver additional effective remedies in the coming era for cancer patients. The objective of this review is to familiarize the reader with the TTP proteins, focus on efficient properties that endow them with their effective oncogenic potential, describe their physiological role in cancer cells, and review the unique properties of TT, and of TTP-driven cancer.
Article
Malignant gliomas are aggressive primary neoplasms that originate in the glial cells of the brain or the spine with notable resistance to standard treatment options. We carried out the study with the aim to shed light on the sensitization of resveratrol to temozolomide (TMZ) against glioma through the Wnt signaling pathway. Initially, glioma cell lines with strong resistance to TMZ were selected by 3‐(4,5‐dimethylthiazol‐2‐yl)‐2,5‐diphenyltetrazolium bromide (MTT) assay. Then, the glioma cells were subjected to resveratrol, TMZ, Wnt signaling pathway inhibitors, and activators. Cell survival rate and inhibitory concentration at half maximum value were detected by MTT, apoptosis by flow cytometry, and terminal deoxynucleotidyl transferase‐mediated dUTP nick‐end labeling staining, in vitro proliferation by hanging drop method and β‐catenin translocation into nuclei by TOP/FOP‐FLASH assay. The expressions of the Wnt signaling pathway‐related and apoptosis‐related factors were determined by western blot analysis. Nude mice with glioma xenograft were established to detect tumorigenic ability. Glioma cell lines T98G and U138 which were highly resistant to TMZ were selected for subsequent experiments. Resveratrol increased the efficacy of TMZ by restraining cell proliferation, tumor growth, and promoting cell apoptosis in glioma cells. Resveratrol inhibited Wnt2 and β‐catenin expressions yet elevated GSK‐3β expression. Moreover, the Wnt signaling pathway participates in the sensitivity enhancing of resveratrol to TMZ via regulating O ⁶‐methylguanine‐DNA methyltransferase (MGMT) expression. Resveratrol sensitized TMZ‐induced glioma cell apoptosis by repressing the activation of the Wnt signaling pathway and downregulating MGMT expression, which may confer new thoughts to the chemotherapy of glioma.
Article
BMI-1 (B-lymphoma Mo-MLV insertion region 1) is a key protein partner in polycomb repressive complex 1 (PRC1) that helps in maintaining the integrity of the complex. It is also a key player in ubiquitination of histone H2A which affects gene expression pattern involved in various cellular processes such as cell proliferation, growth, DNA repair, apoptosis and senescence. In many cancers, Overexpression of BMI1correlates with advanced stages of disease, aggressive clinicopathological behavior, poor prognosis resistance to radiation and chemotherapy. BMI1 is emerging as a key player in EMT, chemo-resistance and cancer stemness. Overexpression is observed in various cancer types such as breast, primary hepatocellular carcinoma (HCC), gastric, ovarian, head and neck, pancreatic and lung cancer. Studies have shown that experimental reduction of BMI protein level in tumor cells results in inhibition of cell proliferation, induction of apoptosis and/or senescence, and increases susceptibility to cytotoxic agents and radiation therapy. Thus, inhibition of BMI1 expression particularly in breast cancer stem cells can be used as a potential strategy for the complete elimination of tumor and to prevent disease relapse. On other hand PTEN is known to be an important tumor suppressor next to p53. In many cancers particularly in breast cancer, p53 and PTEN undergo mutations. Studies have indicated the functional and mechanistic link between the BMI-1oncoprotein and tumor suppressor PTEN in the development and progression of cancer. The current review focuses on recent findings of how oncogenicity and chemo-resistance are caused by BMI1. It also highlights the transcriptional regulation between BMI1 and PTEN that dictates the therapeutic outcome in cancers where the functional p53 is absent. Herein, we have clearly demonstrated the regulation of transcription at genomic loci of BMI1 and PTEN in cancerous tissue or cells and the possible epigenetic regulation by histone deacetylase inhibitors (HDACi) at BMI1 and PTEN loci that may provide some clue for the possible therapy against TNBC in near future.
Article
The histone acetylation modifications (HAMs) influence a large number of cellular functions. They are mediated through histone acetyltransferase (HAT) and histone deacetylase (HDAC). Nowadays, people have realized that HAMs are crucial for development and prognosis of breast cancer. Investigations about abnormal HAMs in breast cancer focus on initiating molecular mechanisms in breast cancer development, identification of new biomarkers to predict breast cancer aggressiveness and the therapeutic potential. As HAMs are reversible, breast cancer may be treated by restoring HAMs to normal levels. Indeed, some HDAC inhibitors have been approved by the US Food and Drug Administration to treat certain cancers. Furthermore, HAT inhibitors, HAT activators and HDAC activators may also be used as drugs to treat breast cancer.