You are on page 1of 50

Accepted Manuscript

Title: Dietary phytochemicals for possible preventive and


therapeutic option of uterine fibroids: signaling pathways as
target

Authors: Md. Soriful Islam James H. Segars Mario


Castellucci MD, PhD Pasquapina Ciarmela Ph.D

PII: S1734-1140(16)30297-3
DOI: http://dx.doi.org/doi:10.1016/j.pharep.2016.10.013
Reference: PHAREP 585

To appear in:

Received date: 20-7-2016


Revised date: 3-10-2016
Accepted date: 19-10-2016

Please cite this article as: Md.Soriful Islam, James H.Segars, Mario Castellucci,
Pasquapina Ciarmela, Dietary phytochemicals for possible preventive
and therapeutic option of uterine fibroids: signaling pathways as target,
http://dx.doi.org/10.1016/j.pharep.2016.10.013

This is a PDF file of an unedited manuscript that has been accepted for publication.
As a service to our customers we are providing this early version of the manuscript.
The manuscript will undergo copyediting, typesetting, and review of the resulting proof
before it is published in its final form. Please note that during the production process
errors may be discovered which could affect the content, and all legal disclaimers that
apply to the journal pertain.
1

FULL TITLE: Dietary phytochemicals for possible preventive and therapeutic option of uterine fibroids:

signaling pathways as target

SHORT TITLE: Dietary phytochemicals for uterine fibroids

Md Soriful Islam1, 2, James H. Segars3, Mario Castellucci1*, Pasquapina Ciarmela1, 4*

1
Department of Experimental and Clinical Medicine, Faculty of Medicine, Università Politecnica delle

Marche, Ancona 60020, Italy


2
Biotechnology and Microbiology Laboratory, Department of Botany, University of Rajshahi, Rajshahi

6205, Bangladesh;
3
Howard W. and Georgeanna Seegar Jones Division of Reproductive Sciences, Department of

Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, USA
4
Department of Information Engineering, Università Politecnica delle Marche, Ancona 60131, Italy

*Corresponding author’s:

1) Pasquapina Ciarmela, Ph.D

Department of Experimental and Clinical Medicine,

Faculty of Medicine, Università Politecnica delle Marche, via Tronto 10/a, 60020 Ancona, Italy

Phone: +390712206270, Fax: +390712206087, E-mail: p.ciarmela@univpm.it

2) Mario Castellucci, MD, PhD

Department of Experimental and Clinical Medicine,

Faculty of Medicine, Università Politecnica delle Marche, via Tronto 10/a, 60020 Ancona, Italy

Phone: +390712206086, Fax: +390712206087, E-mail: m.castellucci@univpm.it


2

Abstract

A growing interest has emerged on dietary phytochemicals to control diverse pathological conditions.

Unfortunately, dietary phytochemical research in uterine fibroids is still under construction. Uterine

fibroids/leiomyomas are benign tumors developing from the myometrium of the uterus in premenopausal

women. They may occur in more than 70 % of women, and approximately 25 % of women show

clinically significant symptoms. These include heavy and prolonged menstrual bleeding, pelvic pressure

(urinary frequency, incontinence, and difficulty with urination), pelvic pain, pelvic mass, infertility, and

reproductive dysfunction. Due to lack of medical treatments surgery has been definitive choice for fibroid

management. Moreover, surgery negatively affects women’s quality of life, and its associated cost

appears to be expensive. The molecular mechanism of fibroids development and growth is not fully

elucidated. However, accumulated evidence shows that several signaling pathways, including Smad 2/3,

PI3K/AKT/mTOR, ERK 1/2 and β-catenin are involved in the leiomyoma pathogenesis, indicating that

they could serve as targets for prevention and/or treatment of this tumor. Therefore, in this review, we

discuss the involvement of signaling pathways in leiomyoma development and growth, and introduce

some potential dietary phytochemicals that could modulate those signaling pathways.

Keywords: Dietary phytochemicals, uterine fibroids, signaling pathways, growth factors, medical

treatment
3

Introduction

Uterine fibroids/leiomyomas are benign tumors developing from the myometrium of the uterus in

premenopausal women [1, 2]. They may occur in more than 70 % of women [3], and among these, about

20-50% of women are reported to produce clinically significant symptoms [4]. Leiomyoma associated

symptoms include: heavy and prolonged menstrual bleeding, pelvic pressure (urinary frequency,

incontinence, and difficulty with urination), pelvic pain, pelvic mass, infertility, and reproductive

dysfunction [5]. Surgery has been a definitive treatment of uterine fibroids. Moreover, surgery negatively

affects women’s quality of life, and its associated cost appears to be expensive. For example, in United

States, surgery associated cost of uterine fibroids management is approximately $5.9-34.4 billion annually

[6].

Unfortunately, the current medical treatments are limited and no effective prevention strategies

exist [7]. Moreover, the benefits of medical treatments are tempered by lack of efficacy or serious adverse

side effects. Poor understanding of the precise molecular mechanism of uterine fibroids development and

growth is may be the reason for the limitation of medical treatments. However, in recent years, significant

advances to uncover the molecular mechanisms of uterine fibroid development and growth have been

achieved [1, 8-11]. It is thought that uterine fibroids are monoclonal tumors, and approximately 40-50 %

contain karyotypic or cytogenetic abnormalities [12]. Notably, mediator complex subunit 12 (MED12)

gene mutation has been noted in 70 % of fibroids [13].

Cell signaling is the transfer of information, by which cells perceive and respond to extracellular

stimuli including growth factors, neurotransmitters, and hormones. The signaling cascade starts with

binding of extracellular stimuli to a cell surface receptor. The receptor then activates series of downstream

signaling molecules in the cytoplasm that import signal to the nucleus for subsequent transcription of

target genes. The activation of several signaling pathways, such as Smad 2/3, phosphoinositide 3-kinase

(PI3K), extracellular-signal-regulated kinase 1/2 (ERK1/2), and β-catenin have been reported in

leiomyoma cells [8-11]. They regulate central events (such as inflammatory response, fibrosis,
4

proliferation and angiogenesis) of leiomyoma development and growth. Therefore, signaling pathways

could serve as excellent target for prevention and treatment of uterine fibroids.

Dietary phytochemicals are plant based chemical compounds with disease-preventive properties,

found in cereals, fruits, vegetables, legumes, herbs, spices, nuts, and beverages (such as tea, wine and

beer). They are known to exert therapeutic effects on multiple pathological conditions through modulating

diverse signaling pathways [14-16]. Accumulating evidences indicate that high intake of green vegetables

and fruit seem to have a protective role and associated with reduced risk of uterine fibroids of US and

Italian populations [17, 18]. This result supports the possible use of dietary phytochemicals for the

prevention and/treatment of uterine fibroids. However, phytochemical based research in uterine fibroids is

still under construction. For example, EGCG (epigallocatechin gallate), curcumin, resveratrol,

isoliquiritigenin and genistein are only few dietary phytochemicals that have been partially studied in

uterine leiomyoma [19-26]. They are mostly known for antiproliferative effects, however, their effects on

signaling pathways have not been addressed in leiomyoma cells except curcumin [21] and genistein [27].

Hence, there is much room for future research in the area of phytochemical based studies focusing on

signaling pathways as therapeutic target. Therefore, in this review, we discussed the role of signaling

pathways in leiomyoma development and growth, and introduced 14 dietary phytochemicals (Fig. 1) that

could modulate those signaling pathways.

Health benefits of dietary phytochemicals

The father of medicine, Hippocrates, proclaimed that “Let food be thy medicine and medicine be thy

food” almost 25 centuries ago. However, the relationship between diet and health is yet to explore. The

human diet contains wide variety of plant-based foods that provide essential nutrients for the body.

Besides, plant-based foods possess huge variety of non-nutrient components that offer beneficiary effects

on health.

Accumulated epidemiologic studies indicate an inverse association between diet rich in fruits and

vegetables and the risk of cancers (such as colon, breast, and ovary) [28-30] and other diseases. The major
5

groups of health promoting dietary phytochemicals are phenolics [phenolic acid-gallic acid, stilbene-

resveratrol, flavonoids (flavonol-quercetin, flavones-apigenin, flavanol-catechin, flavanone-naringenin,

anthocyanidin-delphinidin, and isoflavonoid-genistein), lignin-matairecinol, coumarin-warfarin, and

tannins)], alkaloids-caffeine, carotenoid-lycopene, organosulfur compound-sulforaphane, and

phytosterols-sitosterol and stigmasterol [31]. For example, strawberry, a major source of anthocyanins

(i.e. pelargonidin-3-glucoside), flavonols (quercetin and kaempferol), ellagitannin (ellagic acid), flavanols

(catechins and procyanidins), and phenolic acid (caffeic acid), is known to exert therapeutic effects in

the prevention of diabetes, obesity, cancers, cardiovascular diseases, and neurodegenerative diseases [32].

It is thought that the health benefits of fruits and vegetables are the result of additive and synergistic

interactions of the phytochemicals present in whole foods [33].

Chemoprevention is characterized by the use of natural or synthetic agents to block, reverse or

restrict tumorigenic steps: initiation, promotion, and progression [16]. In recent years, dietary

phytochemicals are being considered as a cost effective, acceptable and accessible approach for cancer

prevention and treatment. Therefore, dietary phytochemicals have been extensively investigated for their

multiple therapeutic effects as well as their safety, and low toxicity [34].

Signaling pathways in uterine leiomyoma

Accumulated evidence suggests that several signaling pathways, including Smad 2/3 (Fig.2A), PI3K

(Fig.2B), ERK 1/2 (Fig.3A), and β-catenin (Fig.3B) activated by growth factors, other peptide or

proteins, and hormones have been found in leiomyoma cells [8-11]. They regulate inflammatory response,

fibrosis, proliferation and angiogenesis that are important phenomenon for leiomyoma pathogenesis.

Since dietary phytochemicals may exert multiple therapeutic effects against diverse pathological

conditions, they could regulate leiomyoma development and growth through targeting signaling

pathways.

Smad 2/3 signaling


6

Smads are group of intracellular proteins that deliver extracellular signal to the nucleus induced by

transforming growth factor (TGF)-β superfamily members. Smads are classified into three different

groups, including receptor-regulated Smads (R-Smad), common-mediator Smad (Co-Smad), and

inhibitory Smads (I-Smad). Smad1, Smad2, Smad3, Smad5 and Smad8 are known as R-Smad. R-Smads

interact with Co-Smad (Smad4) to promote downstream signaling [35]. In contrast, I-Smads (Smad6 and

Smad7) act as blocker of activation of R-Smads and Co-Smads [36].

In myometrial and leiomyoma cells, activin-A and TGF-β1 has been reported to induce Smad 2/3

signaling [9, 37]. They initiate signaling by binding to a type II receptor (ActRIIA or ActRIIB for activin-

A, and TGF-βRII for TGF-β), which recruits and phosphorylates a type I receptor [activin receptor-like

kinase (ALK)-4/ActRIB for activin-A, and ALK5/TGF-βRI for TGF-β). Next, activated type-I receptor

phosphorylates Smad2 and Smad3 that interact with Smad4 in the cytoplasm. This smad complex then

travels to the nucleus where interacts with other transcription factors and regulates transcription of target

genes [38, 39].

Uterine leiomyoma demonstrated an elevated level of Smad3, Smad4 and phosphorylated Smad3

(p-Smad3) as well as TGF-βRI and TGF-βRII expression compared to normal counterpart [40]. TGF-β1

was reported to increase p-Smad3 induction in both myometrial and leiomyoma smooth muscle cells [37].

A number of studies reported that TGF-β1 can modulate inflammatory response, fibrosis, cell growth, and

apoptosis in myometrial and leiomyoma cells [41-45], which may be acquired , at least in part, through

activation of Smad 2/3 signaling [37]. The involvement of Smad 2/3 signaling in leiomyoma growth was

further documented by the observation that TGF-βRI kinase inhibitor, SB-525334 can block TGF-β

signaling in uterine leiomyoma cells, and significantly decreased tumor (leiomyoma) size, incidence and

multiplicity in Eker rat model [46]. Recently, we found that activin-A can induce phosphorylation of

Smad 2/3 in both primary myometrial and leiomyoma cells [9]. The ability of activin-A to increase

mRNA expression of fibronectin, collagen1A1, versican and vascular endothelial growth factor (VEGF)-

A in primary myometrial and/or leiomyoma cells [9, 47], demonstrating its profibrotic and angiogenic

role in this cell types. Ulipristal acetate is one of the most promising therapeutic options for leiomyoma
7

was found to decrease activin-A, follistatin, ActRIIB, and ALK4 mRNA expression in leiomyoma

cultured cells [47]. We also found that ulipristal acetate can block activin-A-induced mRNA expression

of fibronectin and VEGF-A in myometrial and leiomyoma cultured cells [47].

PI3K signaling

PI3Ks are a large family of intracellular signal transducers. Activation of PI3K signaling can occur

through G protein-coupled receptors and tyrosine kinase receptors [48]. Induction of PI3K signaling by

prolactin-releasing peptide (PrRP) and epidermal growth factor (EGF) has been reported in uterine

leiomyoma cells [8, 49]. Upon ligand binding, receptor complex becomes activated, and phosphorylates

PI3K at the cell membrane [48]. Phosphorylated PI3K converts PIP2 (phosphatidylinositol-4,5-

diphosphate) to PIP3 (phosphatidylinositol-3,4,5-triphosphate) [48, 50], which subsequently mediates the

phosphorylation of protein kinase B (PKB/AKT) through phosphoinositide dependent protein kinase 1

(PDK1) [51]. Next, phosphorylated AKT activates/inactivates series of downstream proteins to facilitate

translation of target proteins. mTOR (mammalian target of rapamycin) is a best studied downstream

substrate of AKT. There are two complexes of mTOR: TORC1-Raptor complex and TORC2-Rictor

complex. AKT activates mTOR through phosphorylating and inactivating TSC 1/2, which inhibits mTOR

through GTP-binding protein, Rheb (Ras homolog enriched in brain). Thus, Rheb accumulates and

activates the mTOR-raptor kinase complex. The activated TORC1-Raptor complex mediates

phosphorylation of downstream substrates, eukaryotic translation initiation factor 4E-binding protein 1

(4E-BP1) and p70S6Kinase, which subsequently promote synthesis of target proteins [52].

Accumulated evidences suggest a central role of PI3K/AKT/mTOR pathway in the pathogenesis

of uterine leiomyomas [8, 53-55]. Using in vivo and in vitro studies, Crabtree and co-workers confirmed

the upregulation of mTOR signaling pathway in both human and rat leiomyomas/tumors [53]. They found

that rapamycin analogue WAY-129327 decreased tumor size, incidence and multiplicity in Eker rats, and

inhibited mTOR signaling [53]. MK-2206, an AKT Inhibitor, was reported to reduce mTOR and p70S6K

phosphorylation and promote leiomyoma cell death [56]. Varghese’s group showed that PrRP promoted
8

PI3K/AKT/mTOR pathway through activation of GPR10, and increased primary leiomyoma cell

proliferation [8]. EGF has been reported to increase cell proliferation, and induce phosphorylation and

activation of EGFR and AKT in leiomyomal smooth muscle cells [49]. Involvement of PI3K/AKT

pathway in leiomyoma growth was further evidenced by the observation that inhibition of AKT using

API-59 (AKT inhibitor) decreased leiomyoma cell proliferation and cell viability, and promoted apoptosis

[57]. PTEN (phosphatase and tensin homolog) is a negative regulator of PI3K pathway. Uterine

leiomyoma and myometrium demonstrated an unchanged expression level of PTEN [55].

ERK 1/2 signaling

ERK 1/2 is a cytoplasmic protein [58] that mediates signaling by EGF, platelet-derived growth factor

(PDGF), insulin-like growth factor (IGF)-I and TGF-β in myometrial and/or leiomyoma cells [10, 44,

59]. The signaling is initiated by binding of ligands to their corresponding receptors. Upon lingand

binding, receptor complex (homo- or heterodimers) become auto-and transphosphorylates and activates

that leads to the association of the receptor to cytoplasmic target proteins. Grb2 (growth factor receptor-

bound protein 2 adapter protein), a docking protein, that binds to the activated receptors either directly or

through Shc (src homology and collagen domain protein) [60]. Shc and Grb2 then recruit SOS (son of

sevenless), a guanine nucleotide exchange factor, which in turn activates Ras by exchanging GDP for

GTP. Ras is a small GTP-binding protein that recruits Raf and activates it. Activated Raf then

phosphorylates and activates MEK 1/2, which in turn phosphorylates and activates ERK 1/2 [61].

Activated ERK 1/2 moves to the nucleus where it regulates transcription of target genes [62, 63].

EGF has been reported to stimulate proliferation of leiomyomal smooth muscle cells through

activation of the EGFR-ERK1/2 pathway [49]. Stimulation of primary leiomyoma cells with EGF

markedly increased intracellular reactive oxygen species (ROS) production that mediates mitogen-

activated protein kinase (MAPK)3/MAPK1 (ERK 1/2) activation leading to cell proliferation [10]. The

role of EGF in leiomyoma growth was also supported by the observation that AG1478 and TKS050

(selective EGFR blockers) can block leiomyoma cell proliferation by inducing cell cycle arrest and
9

apoptosis [64, 65]. PDGF has been reported to activate ERK 1/2 signaling through intracellular ROS

production, and increase primary leiomyoma cell proliferation [10]. PDGF also increased collagen α1 (I)

mRNA expression in both myometrial and leiomyoma cells [66]. IGF-I was reported to increase uterine

leiomyoma cell proliferation by upregulating proliferating cell nuclear antigen (PCNA) expression and

downregulating apoptosis by upregulation of B-cell lymphoma-2 (Bcl-2) protein expression [59, 67]. The

stimulatory effect of IGF-I on leiomyoma growth was mediated, at least in part, by increasing

phosphorylation of IGF-IRβ, Shc and MAPKp44/42 (ERK1/2) [59]. TGF-β1 has been shown to regulate

inflammatory response, fibrosis, cell growth, and apoptosis in myometrial and leiomyoma cells [41-45], at

least in part, by ERK 1/2 signaling activation [44].

β-catenin signaling

β-catenin is the central component of wingless-type (WNT) signaling cascade. In the absence of WNT

ligands, β-catenin is degraded by a multiprotein ‘destruction complex’ in the cytoplasm. This

‘destruction complex’ contains APC (adenomatous polyposis coli) and AXIN, which facilitate the

phosphorylation of β-catenin by CK1 (casein kinase 1) and GSK3 (glycogen synthase kinase 3) [68]. In

the presence of WNT ligands, WNT binds to the Frizzled receptors and several co-receptors such as LRP-

5/6 (lipoprotein receptor-related protein-5/6), RYK (receptor-like tyrosine kinase) or ROR2 (receptor

tyrosine kinase-like orphan receptor 2) [69] resulting in the inhibition of ‘destruction complex’ that

stabilize β-catenin in the cytoplasm. Next, stabilized β-catenin moves to the nucleus and interacts with

LEF (lymphoid enhancer factor)/TCF (T-cell factor) transcription factors to regulate transcription of

target genes [70].

A considerable amount of evidence has suggested that WNT/β-catenin signaling is involved in

the pathogenesis of uterine fibroids [11, 71-73]. Tanwar and co-investigators developed a mouse model

that expresses constitutively activated β-catenin in uterine mesenchyme which gives rise to mesenchymal

tumors/leiomyoma-like tumors in the uterus [71]. The constitutive activation of β-catenin induced the

expression of TGF-β3 [71], which was shown to induce proliferation and extracellular matrix (ECM)
10

formation in human uterine leiomyoma cells [74]. The tumor suppressor gene, MED12, is mutated in ~

70% of uterine leiomyomas [13], which directly binds to β-catenin and regulates WNT signaling [75].

The expression of WNT4 was found to be elevated in leiomyoma with MED12 mutations versus those

without mutations [73]. An elegant experiment by Ono and co-workers uncovered a central role of the

WNT/β-catenin pathway induced by estrogen and progesterone in leiomyoma growth, which was

confirmed by the observation that blocking of WNT activity through inhibitor of β-Catenin and TCF4

inhibits the growth of leiomyoma-like tumors in immunodeficient mice [11]. They found that WNT11

and WNT16 were increased at mRNA levels in myometrial cells, which were remained constitutively

increased (not significant) in leiomyoma cells in response to estrogen and progesterone treatment [11]. In

addition, frizzled receptors, FZD1 and FZD7 were found to be significantly higher at mRNA levels in

leiomyoma side-population cells than in total leiomyoma cells or leiomyoma mature population cells [11].

This group also found that LRP5 and LRP6 (co-receptors for WNT) were expressed in leiomyoma side-

population cells and leiomyoma mature population cells. Furthermore, estrogen and progesterone were

found to selectively induce nuclear translocation of β-catenin and transcriptional activity of TCF and

AXIN2 in leiomyoma side-population cells cocultured with mature myometrial cells [11].

Dietary phytochemicals that might target signaling pathways in uterine leiomyoma

Here, we discuss 14 dietary phytochemicals that might target signaling pathways, including Smad 2/3

(Fig.2A), PI3K (Fig.2B), ERK 1/2 (Fig.3A), and β-catenin (Fig.3B) involved in cell proliferation,

angiogenesis, inflammation and fibrosis that are linked to uterine leiomyoma development and growth.

These dietary phytochemicals were chosen based on their ability to modulate signaling pathways in

different pathological cell types.

Betulinic acid

Betulinic acid is a pentacyclic triterpene found in leaves of rosemary, and fruits of elephant apple.

Recently, Jin and co-workers reported that betulinic acid can inhibit 3-isobutyl-1-methylxanthine induced
11

melanogenesis via the downregulation of p-MEK, p-ERK and p-AKT in B16F10 cells [76]. Betulinic

acid may exert antiangiogenic activity in cultured endometrial adenocarcinoma cells through decreasing

expression of hypoxia-inducible factor (HIF)-1α, and VEGF [77]. In addition, betulinic acid was reported

to suppress lipopolysaccharide (LPS)-induced interleukin (IL)-6 production through preventing nuclear

translocation of nuclear factor-κB (NF-κB) in peripheral blood mononuclear cells [78]. The antifibrotic

effect of betulinic acid was documented by the observation that betulinic acid can attenuate liver hepatic

stellate cell (HSC) activation by downregulating ethanol-induced p-Smad3 expression [79].

Butein

Butein is a type of chalcone derivative found in cashews. Khan and co-workers reported that butein can

inhibit prostate tumor growth in vivo through inhibition of PI3K/AKT pathway [80]. Butein also

suppressed breast cancer cell growth through downregulation of AKT phosphorylation [81]. Furthermore,

butein was reported to inhibit cell proliferation and clonogenecity of bladder cancer cells, through

reducing phosphorylation of ERK 1/2 [82].

Butein has been reported to inhibit VEGF-induced angiogenesis by downregulating the

phosphorylation of AKT, mTOR, and the major downstream effectors, p70S6K, 4E-BP1, and eIF4E in

endothelial progenitor cells [83]. Matrix metallopeptidase (MMP)-9 and VEGF are prominently involved

in the processes of tumor cell invasion and metastasis. It was shown that butein repressed tumor necrosis

factor (TNF)-α and phorbol-12-myristate-13-acetate induced expression of VEGF and MMP-9 via the

suppression of NF-κB activity in human prostate cancer cells [84].

The anti-inflammatory effect of butein has been reported by several investigators [85-87]. Wang

and co-workers reported that butein suppressed adipocyte inflammation in 3T3-L1 cells by

downregulation of TNF-α+LPS+interferon (IFN)-γ-induced secretion and/or expression of inflammatory

mediators, such as IL-6, monocyte chemoattractant protein-1 (MCP-1), inducible nitric oxide synthase

(iNOS), nitric oxide (NO), NOS2 (nitric oxide synthase 2), CXCL (C-X-C motif chemokines)-1, and

CXCL-10) through partly suppression of NF-κB activation and MAPKs [ERK 1/2, c-Jun N-terminal
12

protein kinase (JNK), and p38 MAPK] signaling pathways [87]. Butein also decreased TNF-α-induced

monocyte cell adhesion to lung epithelial cells through inhibiting ROS generation and NF-κB activation

as well as the phosphorylation of MAPKs and AKT [85]. Furthermore, butein was reported to ameliorate

colitis in IL-10(-/-) mice, at least in part, by downregulation of IL-6, IL-1β, IFN-γ and MMP-9 as well as

IL-6-induced activation of signal transducer and activator of transcription (STAT)3 [86].

The antifibrotic effect of butein has been documented by the observation that butein can inhibit

ethanol-induced activation of liver stellate cells through inhibition of TGF-β, p38 MAPK, and JNK

signaling pathways [88].

Capsaicin

Capsaicin, active component of chili peppers, is known to have tumor suppressive effects. Recently, Park

and co-workers reported that capsaicin can potentially inhibit the proliferation of human gastric cancer

cells and induce apoptosis, through decreasing the expression of p-ERK 1/2 [89]. Capsaicin was also

reported to exert anticancer effect on human colorectal cancer cells through modulating β-catenin

signaling pathway [90]. This compound promoted proteosomal- degradation of β-catenin, and suppressed

TCF-4 expression and disrupted the interaction between TCF-4 and β-catenin [90].

Capsaicin can induce antiangiogenic activity both in vitro and in vivo [91]. Treatment of human

multiple myeloma cells with capsaicin inhibited IL-6-induced STAT3 activation, and STAT3-regulated

gene products, such as cyclin D1, Bcl-2, Bcl-xL, survivin, and VEGF [92], suggesting its regulatory

function in proliferation, survival and angiogenesis, and apoptosis. In addition, capsaicin was reported to

increase degradation of HIF-1α, which is a key transcription factor in increasing VEGF transcription in

non-small cell lung carcinoma [93]

Bitencourt and co-investigators reported that capsaicin induced down-regulation of HSC

activation by decreasing cyclooxygenase (COX)-2, TGF-β1 and collagen expression [94], suggesting its

antifibrotic and antiinflammatory actions. Accordingly, capsaicin has been reported to suppress the
13

production of TNF-α [95], as well as prostaglandin E2 (PGE2) and NO production in macrophages via

NF-κB inactivation [96].

Delphinidin

Delphinidin is a polyphenolic compound found in many pigmented fruits, including cranberries, Concord

grapes, and pomegranates. A recent study reported that delphinidin suppressed proliferation and migration

of human ovarian clear cell carcinoma cells through blocking phosphorylation of downstream targets of

PI3K (AKT and p70S6K) and MAPKs (ERK1/2 and JNK) [97]. Pal’s group demonstrated that

delphinidin can inhibit growth of non-small-cell lung cancer cells through inhibition of EGFR, VEGFR2

as well as PI3K/AKT and MAPKs pathways [98]. Delphinidin also inhibited in vivo tumor growth

induced by B16-F10 melanoma cell xenograft in mice, through suppression of VEGFR2-mediated

endothelial cell proliferation as well as VEGFR2 signaling pathways, MAPKs (ERK1/2 and p38 MAPK)

and PI3K [99].

Angiogenesis is an energetic process and VEGF-induced angiogenesis is associated with

mitochondrial biogenesis. Delphinidin was reported to inhibit VEGF induced-mitochondrial biogenesis

and AKT activation in endothelial cells [100]. Lamy and co-workers reported that delphinidin can inhibit

smooth muscle cell migration as well as the differentiation and stabilization of endothelial cells, at least in

part, by inhibiting activation of PDGFR, and PDGF-BB-induced activation of ERK-1/2 signaling

pathway [101].

In athymic nude mice implanted with human prostate cancer PC3 cells, delphinidin treatment

induced a significant inhibition of tumor growth, through downregulation of NF-κB expression [93].

Delphinidin also inhibited UVB-induced MMP-1 expression as well as ROS production and NOX activity

in primary cultured human dermal fibroblasts partly through suppression of MAPKs phosphorylation

[102].

Delphinidin has been reported to inhibit TGF-β1-induced expression of α-smooth muscle actin

(α-SMA), fibronectin, and collagen as well as activation of MAPKs and NF-κB in nasal polyp-derived
14

fibroblasts [103], suggesting its role in the regulation of myofibroblast differentiation and ECM

production through the MAPK/NF-κB signaling pathway.

3,3′-Diindolylmethane

3,3′-Diindolylmethane (DIM) is a polymeric compound produced after digestion of indole-3-carbinol in a

low pH environment. Several cruciferous vegetables, such as Brussels sprouts, cauliflower, cabbage,

broccoli, kale and turnips are known to be major sources of DIM. It has been shown that DIM can induce

antiproliferative and proapoptotic effects in oral squamous cell carcinoma cells and breast cancer cells,

through inactivation of NF-κB, AKT and MAPKs pathways [104, 105]. DIM also induced anti-

proliferative and pro-apoptotic effects in human cervical cancer cells through downregulating the

expression of p-AKT, PI3K, GSK-3β, p-PDK1 as well as p-c-Raf, p-ERK1/2 and p-p38 MAPK [106]. A

recent genome-wide transcriptome analysis showed that DIM can inhibit proliferation of colon cancer

cells through inactivation of Wnt/β-catenin signaling pathway [107]. DIM also suppressed the growth of

ovarian tumors in vitro and in vivo, at least in part, through reduction of EGFR, MEK, and ERK

phosphorylation [108].

Chang and co-workers found that DIM can inhibit VEGF-induced cell proliferation in human

umbilical vascular endothelial cells (HUVECs), at least in part, via downregulation of ERK1/2 and AKT

[109, 110]. DIM also inhibited invasion and angiogenesis in PDGF-D-overexpressing PC3 cells through

inactivation of mTOR and AKT [111].

The anti-inflammatory effect of DIM was documented by the observation that DIM can inhibit

LPS-induced microglial hyperactivation and brain inflammation through attenuating DNA-binding

activity of NF-κB and phosphorylation of inhibitor of κB [112].

Zhang and co-investigators reported that DIM can attenuate hepatic fibrosis by inhibiting miR-21

expression, and TGF-β induced α-SMA and COL1A1 as well as p-Smad2/3 and total Smad2/3 expression

[113]. A recent study reported that DIM can attenuate TGF-β1-induced myofibroblastic transformation of
15

cardiac fibroblast through suppression of α-SMA, collagen I, collagen III, and connective tissue growth

factor (CTGF) expression via downregulation of AKT/GSK-3β signaling pathways [114].

Emodin

Emodin is an anthraquinone compound found in the root and rhizomes of Rhubarb. It has been shown that

emodin can inhibit tumor growth in orthotopic hepatocellular carcinoma mice model, at least in part, by

blocking of STAT3, JAK1/2 and AKT phosphorylation [115]. Emodin also downregulated the expression

of STAT3 regulated gene products, such as cyclin D1, Bcl-2, Bcl-xL, Mcl-1, survivin and VEGF in

HepG2 cells [115]. In human colorectal cancer cells, emodin downregulated the expression of β-catenin

and TCF7L2, as well as several downstream proteins, including cyclin D1, c-Myc, snail, vimentin, MMP-

2 and MMP-9 [116]. Emodin was also reported to repress TWIST1 (Twist-related protein 1)-induced

epithelial-mesenchymal transition (EMT) in head and neck squamous cell carcinoma FaDu cells through

inactivation of β-catenin and AKT signaling pathways [117]. A recent study reported that emodin can

inhibit the TGF-β-induced migration and invasion of human cervical cancer cells, partly through

decreasing the expression of TGF-βRII, p-Smad3, Smad4, and β-catenin [118].

Kaneshiro and co-investigators reported that emodin can inhibit endothelial cell proliferation,

migration, and tube formation through blocking ERK 1/2 phosphorylation [119], suggesting its

antiangiogenic properties.

Emodin has been reported to inhibit homocysteine-induced C-reactive protein generation in

vascular smooth muscle cells, through downregulating ROS generation, and phosphorylation of ERK1/2

and p38 MAPK [120], suggesting its antiinflammatory and antiatherosclerotic effects. Yin and co-

investigators reported that emodin can protect against LPS/D-galactosamine-induced liver injury in mice

through attenuating TNF-α production as well as p38 MAPK and NF-κB activation [121]. In addition,

emodin was reported to ameliorate LPS-induced mastitis in mice through reducing secretion and

expression of TNF-α, IL-1β and IL-6 via inactivating NF-κB and MAPKs pathways [122].
16

Emodin can exhibit antifibrotic effect on pancreatic fibrosis, at least in part, by reducing collagen

and TGF-β1 expression (Wang et al., 2007a). Lee and co-workers reported that emodin suppressed TNF-

α-induced MMP-1 expression in human dermal fibroblast cells through inhibition of the activator protein

1 (AP-1) and MAPKs (ERK 1/2 and JNK) pathways [123]. Emodin was also reported to suppress

glucose/IL-1β induced mesangial cell proliferation and ECM (fibronectin and/or collagen) production by

blocking p38 MAPK pathway [124, 125].

Ferulic acid

Ferulic acid is a ubiquitous polyphenolic compound in plant kingdom, found especially in artichokes,

eggplants and maize bran. Ambothi and co-investigators reported that ferulic acid can inhibit UVB-

radiation induced photocarcinogenesis through downregulation of VEGF, iNOS, mutant p53, Bcl-2

expressions and upregulation of the Bax expression [126], suggesting its antiinflammatory,

antiangiogenic and apoptosis inducing capability.

A recent report indicated that ferulic acid can inhibit fibroblast growth factor (FGF)1-induced

endothelial cell proliferation, migration and tube formation as well as microvessel sprouting of rat aortic

rings and angiogenesis [127]. The antiangiogenic effect of ferulic acid was mediated by inactivation of

FGFR1 and PI3K/PKB signaling [127]. Hou’s group reported that ferulic acid suppressed proliferation of

ECV304 endothelial cells and blocked the cell cycle in G0/G1 phase, and inhibited phosphorylation of

ERK 1/2 [128].

Recently, Xu and co-workers reported that ferulic acid can exhibit antifibrotic effects on hepatic

stellate cells through inhibiting ERK 1/2 and Smad 2/3 signaling pathways [129]. Particularly, this

compound reduced TGF-βRII, TGF-βRI and Smad4 mRNA and protein expression as well as Smad

transcriptional activity, and blocked ERK 1/2 phosphorylation in HSC-T6 cells [129]. Ferulic acid also

attenuated TGF-β1-induced renal cellular fibrosis in NRK-52E cells via inactivation of Smad 2/3

signaling pathway [130]. Furthermore, ferulic acid was reported to attenuate ischemia/reperfusion-

induced hepatocyte apoptosis via inhibition of JNK activation [131].


17

Fisetin

Fisetin is a flavonoid commonly present in apples, kiwis, strawberries, grapes, persimmons, onions and

cucumbers. Khan and co-workers reported that fisetin can inhibit growth of human non-small cell lung

cancer cells via suppression of PI3K/AKT/mTOR signaling [132]. In PC3 prostate cancer cells, fisetin

induced autophagic cell death through inhibition of mTOR signaling pathway [133].}. Particularly, fisetin

inhibited phosphorylation of mTOR kinase and downregulated the expression of Raptor, Rictor, PRAS40

and GβL as well as activated the mTOR repressor, TSC2 in this cell type [133]. Furthermore, fisetin was

reported to inhibit human laryngeal carcinoma cells of TU212 cell proliferation and induce apoptosis via

downregulating Raf, Ras, p-ERK1/2, PI3K, p-AKT, NF-κB and mTOR expression [134].

Fisetin has been reported to exert antiangiogenic effect by inhibiting VEGF-induced growth and

survival of HUVEC as well as capillary-like tube formation on Matrigel [135]. It also inhibited the

expression of eNOS (endothelial nitric oxide synthase), VEGF, iNOS, MMP-2 and MMP-9 in A549 and

DU145 human cancer cells [135].

Fisetin exerts antiinflammatory effects in human mast cells by suppressing phorbol-12-myristate

13-acetate plus calcium ionophore A23187-stimulated gene expression and production of TNF-α, IL-1β,

IL-4, IL-6, and IL-8 via downregulation of MAPKs and NF-κB pathways [136]. Fisetin also reduced

secretion of IL-6 and TNF-α via inactivation of JNK and NF-κB in LPS-stimulated macrophage cells

[137]. Furthermore, fisetin was reported to inhibit IL-1β-induced cytokines (TNF-α, IL-6) and

chemokines (IL-8, MCP-1) in rheumatoid arthritic fibroblast-like synovial cells and in vivo models [138].

Kaempferol

Kaempferol is a flavonoid present in green tea, broccoli, apples, strawberries and green beans. A recent

study reported that kaempferol can suppress estrogen and triclosan stimulated breast cancer cell growth in

cellular and xenograft breast cancer models via downregulation of p-AKT and p-MEK1/2 expression

[139]. Kaempferol also inhibited proliferation of esophageal squamous cell carcinoma cells and induced
18

G0/G1 cell cycle arrest, and suppressed tumor growth in KYSE150 xenograft model via suppression of

EGFR and its downstream signaling pathways, ERK 1/2 and AKT [140]. Furthermore, kaempferol was

reported to suppress bladder cancer tumor growth by inhibiting cell proliferation and inducing apoptosis

via downregulation of the p38 MAPK phosphorylation and c-Fos expression [141]. Kim and co-workers

reported that kaempferol inhibited PDGF-BB-induced rat aortic vascular smooth muscle cell proliferation,

at least in part, by downregulation of PDGFR-β phosphorylation and its downstream signal transduction

pathways, ERK1/2, AKT and PLC-γ1 phosphorylation and c-fos expression [142]. The ability of

kaempferol to downregulate IGF-I-induced phosphorylation of the IGF-IR and ERK 1/2 pathways in HT-

29 human colon cancer cells was reported by Lee and co-investigators [143].

In chorioallantoic membranes of chicken embryos, kaempferol inhibited OVCAR-3-induced

angiogenesis and tumor growth, at least in part, by reducing VEGF and HIF-1α expression via

downregulation of AKT phosphorylation [144]. Kaempferol also suppressed hepatocarcinoma cell

survival through downregulation of HIF-1 activity and p44/42 MAPK activation [145]. Furthermore,

kaempferol was reported to inhibit VEGF secretion, and in vitro angiogenesis, via downregulation of

ERK phosphorylation as well as NF-κB and cMyc expression [146].

Kaempferol exhibited a protective effect on LPS-induced acute lung injury in mice via

suppression of TNF-α, IL-1β and IL-6 production and MAPKs and NF-κB phosphorylation [147].

Kaempferol also attenuated myocardial ischemic injury via downregulation of TNF-α and IL-6

production as well as inhibition of MAPKs phosphorylation and NF-κB expression [148]. Yoon and co-

investigators reported that kaempferol can inhibit IL-1β-induced proliferation of rheumatoid arthritis

synovial fibroblasts and the expression of COX-2, PGE2 as well as MMP-1 and MMP-3, partly through

downregulation of NF-κB activation as well as MAPKs phosphorylation [149].

The antifibrotic effect of kaempferol was documented by the observation that kaempferol can

suppress collagen deposition, epithelial excrescency and goblet hyperplasia in the lung of ovalbumin-

challenged mice, at least in part, by inhibition of TGF-β-induced EMT by reversing E-cadherin

expression and retarding the induction of N-cadherin and α-SMA [150].


19

Morin

Morin is a member of flavonols found in almonds that suppressed growth and invasion of the metastatic

breast cancer cell line MDA-MB 231 as well as cancer cell progression in xenograft mouse model, partly

through suppression of AKT pathway [151].

Morin has been reported to attenuate ovalbumin-induced airway inflammation in mice by

suppression of goblet cell hyperplasia and collagen deposition and ovalbumin-induced TNF-α, IL-4, IL-

13, and MMP-9 via inactivating MAPKs pathway [152]. In human bronchial epithelial cells, morin also

inhibited TNF-α induced ROS generation and expression of eotaxin-1, MCP-1, and IL-8 [152].

Furthermore, morin was reported to suppress monosodium urate crystal-induced inflammation in RAW

264.7 macrophages through inhibition of expression and/or secretion of inflammatory mediators (TNF-α,

IL-1β, IL-6, MCP-1, NO, PEG2, iNOS and COX-2), VEGF expression, ROS generation, and NF-κB

activation [153].

Morin has been reported to ameliorate in vivo diethylnitrosamine-induced liver fibrosis in male

albino Wistar rat as well as inhibit proliferation of LX-2 cells (culture-activated human HSCs), partly

through inhibition of β-catenin and GSK-3β expression [154].

Naringin

Naringin is a flavanone glycoside found in grapefruit and related citrus species. Li and co-workers

reported that naringin can inhibit proliferation of triple-negative (ER-/PR-/HER2-) breast cancer cells

(MDA-MB-231, MDA-MB-468 and BT-549), through induction of apoptosis and G1 cycle arrest [155].

They also noticed that inactivation of β-catenin signaling pathway was responsible for this anticancer

effect [155]. Naringin also induced autophagy-mediated growth inhibition in AGS cancer cells by

downregulating the phosphorylation of PI3K and its activated downstream targets p-AKT and p-mTOR

via activation of MAPKs pathways [156].


20

Recently, Zhang and co-workers reported that naringin can prevent intestinal tumorigenesis in

Apc (Min/+) mouse model through suppression of cell proliferation, induction of apoptosis and inhibition

of expression and/or secretion of inflammatory mediators (Cox-2, NF-κB, TNF-α, PGE2 and IL-6) as

well as β-catenin expression and GSK-3β phosphorylation [157]. Naringin also inhibited growth of HeLa

cervical cancer cells and induces apoptosis, at least in part, by suppression of NF κB phosphorylation

and COX 2 expression [158]. Furthermore, naringin was reported to inhibit TNF-α-induced invasion and

migration of vascular smooth muscle cells as well as expression and/or secretion of MMP-9, IL-6 and IL-

8, at least in part, through blocking of PI3K/AKT/mTOR/p70S6K pathway via suppression of

transcriptional activity of AP-1 and NF κB [159].

The protective effects of naringin against paraquat-induced acute lung injury and pulmonary

fibrosis in mice have been reported, which was mediated primarily through downregulation of expression

of TGF-β1 and TNF-α as well as modulation of expression and ratios of MMP-9 and TIMP-1 [160].

Naringin also inhibited renal interstitial fibrosis and collagen formation partly through inhibiting

oxidative stress and NF-κB activation [161].

Pterostilbene

Pterostilbene is a stilbene, biologically classified as a phytoalexin, chemically related to resveratrol.

Several types of grapes and blueberries are major sources of pterostilbene. Pterostilbene has been reported

to inhibit 7,12-dimethylbenz[a]anthracene/12-O-tetradecanoylphorbol-13-acetate-induced mouse skin

tumor formation possibly via inactivation of NF-κB and AP-1, and their upstream signaling pathways,

MAPKs, PI3K and AKT [162]. Paul’s group reported that pterostilbene can inhibit the growth of cultured

colon cancer HT-29 cells and induce apoptosis through downregulating TNF-α+IFN-γ+LPS-induced

iNOS and COX-2 expression via suppression of p38 MAPK signaling pathway [163]. Later study

reported that pterostilbene can reduce colon tumor multiplicity of non-invasive adenocarcinomas in rats

by inhibiting TNF-α, IL-1β and IL-4 expression via suppression of β-catenin and NF-κB signaling

pathways [164]. Chiou and co-workers reported that pterostilbene inhibited azoxymethane-induced
21

colorectal aberrant crypt foci and adenomas through upregulation of apoptosis and downregulation of

iNOS, COX-2, VEGF, cyclin D1, MMP-7, MMP-26, MMP-2, and MMP-9 expression and/or activity, at

least in part, via suppression of multiple signaling molecules, including p-GSK-3β, β-catenin, p-PI3K, p-

AKT, EGF, and EGFR [165].

Pterostilbene has been reported to suppress 12-O-tetradecanoylphorbol 13-acetate-induced

invasion, migration and metastasis of HepG2 cells by downregulation of angiogenic factors, such as

VEGF, EGF and EGFR expression, and their downstream signal transduction pathways, MAPKs,

PI3K/AKT and PKC as well as NF-κB and AP-1 [166].

Pterostilbene has been reported to inhibit high fat-induced atherosclerosis inflammation in mice

through inhibition of TNF-α, TGF-β1, IL-18, IL-6, IFN-γ, MCP-1 and IL-17 expression and/or secretion

via inactivation of NF-κB [167]. Pterostilbene also reduced the expression of TNF-α, IL-1 β, IL-6, COX-

2, MMP-2 and MMP-9, and ROS overproduction in hyperosmotic medium exposed human corneal

epithelial cells [168], suggesting its ability to protect corneal epithelial cells through antiinflammatory and

antioxidative effects.

It has been shown that pterostilbene inhibited dimethylnitrosamine-induced liver fibrosis in rats

[169]. The antifibrotic effect of pterostilbene in the dimethylnitrosamine-treated rats was mediated by

downregulation of α-SMA and MMP-2 expression as well as TGF-β1, and its signaling molecules, p-

Smad2 and p-Smad3 expression [169].

Silibinin

Silibinin (also known as silybin) is a flavonolignan compound found artichokes. This compound can

induce antiproliferative, antiinflammatory, proapoptotic and antiangiogenic effects in colorectal cancer as

evidenced by decreased cell proliferation [170, 171], and TNF-α-induced NF-κB activation, and thereby

NF-κB-regulated molecules, including Bcl-2, COX-2, iNOS, VEGF, HIF-1α and MMP9 [170-172]. This

effect was mediated, at least in part, by downregulation of β-catenin, IGF-1Rβ, p-GSK-3β, p-ERK 1/2

and p-AKT [170, 171]. Kim’s group reported that silibinin can suppress EGF-induced phosphorylation of
22

EGFR and ERK 1/2 in SKBR3 and BT474 breast cancer cells [173]. In nude mice model, silibinin also

attenuated the growth of melanoma xenograft tumors through inhibiting phosphorylation of MEK 1/2 and

ERK 1/2 [174].

Antiangiogenic effect of silibinin was documented by the observation that silibinin can inhibit

VEGF secretion and HIF-1α subunit accumulation in retinal pigmented epithelia cells through

downregulation of p-PI3K, p-AKT, p-mTOR and p70S6K [175]. In the rat model of age-related macular

degeneration, silibinin also prevented VEGF- and VEGF plus hypoxia-induced retinal oedema and

neovascularization [175]. Furthermore, silibinin was reported to inhibit human cervical and hepatoma

cancer cell growth by inhibiting HIF-1α protein synthesis and hypoxia-induced VEGF secretion via

suppression of the p-mTOR and its effectors, p70S6K and 4E-BP1 [176].

Silibinin has been reported to suppress LPS-induced neutrophilic airway inflammation, at least in

part, by downregulation of ERK phosphorylation [177]. Silibinin also inhibited ovalbumin-induced

airway inflammation, and reduced the production of various cytokines (TNF-α, IL-1β, IL-4, IL-5, and IL-

13), partly via downregulation of NF-κB activity [178]. Furthermore, silibinin showed inhibitory effect on

skin inflammation induced by 12-O-tetradecanoylphorbol-13-acetate by down-regulating IL-1β, IL-6,

TNF-α and COX-2 expression via inhibiting the PI3K/AKT signaling pathway, and NF-κB activation

[179].

Silibinin has been reported to attenuate cardiac hypertrophy and fibrosis by downregulating

EGFR-dependent ERK1/2 and PI3K/AKT, as well as activation of NF-κB and Smad 2/3 signaling

pathways [180]. Chen and colleagues reported that silibinin can inhibit myofibroblast transdifferentiation

in human tenon fibroblasts and reduce fibrosis in a rabbit trabeculectomy model [181]. Particularly,

silibinin inhibited TGF-β1-induced expression of α-SMA, vimentin, collagen contraction, CTGF,

collagen type I in human tenon fibroblasts through downregulation of p-Smad3 [181]. A study by Cho

and co-workers indicated that silibinin has the potential to prevent fibrotic skin changes by inducing the

downregulation of type I collagen expression in human skin fibroblasts, partly by the inhibition of TGF-

β1-induced p-Smad2 and p-Smad3 expression [182]. Silibinin exerts antiinflammatory and antifibrogenic
23

effects on human hepatic stellate cells by downregulation of human HSC cell proliferation, cell migration,

and de novo synthesis of collagen type I as well as IL-1β-induced synthesis of MCP-1 and IL-8 via

directly inhibiting ERK, MEK, and Raf phosphorylation [183].

Thymoquinone

Thymoquinone is a bioactive component of black seed oil. It has been shown that thymoquinone can

induce cell cycle arrest and apoptosis in breast cancer cells through inhibition of PI3K/AKT pathway

[184, 185]. Particularly, thymoquinone reduced the phosphorylation of PTEN (inactivated form of

PTEN), PDK1 and AKT that resulted in the inhibition of 4E-BP1 and p70S6K [184]. In human

cholangiocarcinoma cells, thymoquinone induced inhibition of cell growth by inducing cell cycle arrest

and apoptosis through downregulation of PI3K/AKT and NF-κB pathways, and their regulated gene

products, including Bcl-2, COX-2, and VEGF [186]. Furthermore, thymoquinone was reported to inhibit

proliferation and invasion of human nonsmall-cell lung cancer cells via downregulation of ERK pathway

as confirmed by the reduced expression level of PCNA, cyclin D1, MMP-2 and MMP-9, and p-

ERK1/2[187].

Thymoquinone has been reported to prevent tumor angiogenesis in a mouse xenograft human

prostate cancer (PC3) model, and inhibited human prostate tumor growth through suppressing AKT and

ERK signaling pathways [188]. Thymoquinone also exhibited antiangiogenic effects on osteosarcoma in

vitro and in vivo through inactivating the NF-κB pathway as evidenced by downregulation of NF-κB

DNA-binding activity, survivin and VEGF in SaOS-2 cells [189].

A recent study by Su and co-investigators reported that thymoquinone can inhibit inflammation

and neoangiogenesis induced by Ovalbumin in asthma mice by reducing IL-4 and IL-5 production, as

well as VEGF, p-VEGFR2, p-PI3K and p-AKT expression and tube information in HUVECs [190].

Thymoquinone also inhibited IL-1β-induced inflammation in human osteoarthritis chondrocytes as

evidenced by decreased production of COX-2, iNOS, NO, and PGE2 as well as MMP-1, MMP-3, and

MMP-13 expression via inhibiting NF-κB activation and IκBα degradation as well as MAPKs pathway
24

activation [191]. Furthermore, thymoquinone was reported to inhibit TNF-α-induced IL-6 and IL-8

production in rheumatoid arthritis synovial fibroblasts by inhibiting JNK and p38 MAPK signaling

pathways [192].

Thymoquinone has been reported to attenuate liver fibrosis by reducing α-SMA, collagen1A1,

collagen3A1, TIMP-1, L-1α, IL-1β and IL-18 levels, at least in part, via inactivating PI3K and TLR4

signaling pathways [193-195]. Thymoquinone can also block lung injury and fibrosis in rats induced by

bleomycin/paraquat herbicide through downregulation of TGF-β1, α-SMA, collagen 1A1 and collagen

4A1, and inhibition of oxidative stress and NF-κB activation [196, 197].

Conclusions and future perspectives

The precise molecular mechanisms of leiomyoma pathogenesis are not well understood. However,

accumulating evidences suggest that several signaling pathways, such as Smad 2/3, PI3K, ERK1/2 and β-

catenin are involved in regulating central events (such as inflammatory response, fibrosis, proliferation

and angiogenesis) of leiomyoma development and growth. Thus, they may act as excellent target for

possible prevention and treatment of uterine fibroids. Here, we introduced 14 dietary phytochemicals

(betulinic acid, butein, capsaicin, delphinidin, 3,3'-diindolylmethane, emodin, ferulic acid, fisetin,

kaempferol, morin, naringin, pterostilbene, silibinin and thymoquinone) that could potentially be used as

therapeutic and/or preventive compounds for uterine leiomyoma (Fig. 1). These dietary phytochemicals

have shown their ability to regulate major tumor initiating and promoting events such as, inflammation,

fibrosis, proliferation and angiogenesis in different experimental conditions through regulating several

signaling pathways, including Smad 2/3, PI3K, ERK 1/2 and β-catenin. Future research may include these

dietary phytochemicals to check their ability to modulate signaling pathways in uterine fibroids. Dietary

phytochemicals are not limited to these 14 compounds. Other potential dietary phytochemicals that have

not yet been tested could be included for future research in uterine fibroids.
25

Funding body

This work was supported by a grant from the “Fondazione Cassa di Risparmio di Fabriano e

Cupramontana” (to MC and PC).

Conflict of interest

None

References

[1] Islam MS, Protic O, Stortoni P, Grechi G, Lamanna P, Petraglia F, et al. Complex networks of

multiple factors in the pathogenesis of uterine leiomyoma. Fertil Steril. 2013;100:178-93.

[2] Bulun SE. Uterine fibroids. N Engl J Med. 2013;369:1344-55.

[3] Day Baird D, Dunson DB, Hill MC, Cousins D, Schectman JM. High cumulative incidence of uterine

leiomyoma in black and white women: ultrasound evidence. Am J Obstet Gynecol. 2003;188:100-7.

[4] Buttram Jr VC, Reiter RC. Uterine leiomyomata: etiology, symptomatology, and management. Fertil

Steril. 1981;36:433-45.

[5] Sabry M, Al-Hendy A. Innovative oral treatments of uterine leiomyoma. Obstet Gynecol Int.

2012;2012:943635.

[6] Cardozo ER, Clark AD, Banks NK, Henne MB, Stegmann BJ, Segars JH. The estimated annual cost

of uterine leiomyomata in the United States. Am J Obstet Gynecol. 2012;206:211. e1-. e9.

[7] Islam MS, Protic O, Toti P, Giannubilo SR, Tranquilli AL, Petraglia F, et al. Uterine leiomyoma:

available medical treatments and new possible therapeutic options. J Clin Endocrinol Metab.

2013;98:921–34.

[8] Varghese BV, Koohestani F, McWilliams M, Colvin A, Gunewardena S, Kinsey WH, et al. Loss of

the repressor REST in uterine fibroids promotes aberrant G protein-coupled receptor 10 expression and

activates mammalian target of rapamycin pathway. Proc Natl Acad Sci U S A. 2013;110:2187-92.
26

[9] Islam MS, Catherino WH, Protic O, Janjusevic M, Gray PC, Giannubilo SR, et al. Role of activin-A

and myostatin and their signaling pathway in human myometrial and leiomyoma cell function. J Clin

Endocrinol Metab. 2014;99:E775-E85.

[10] Mesquita FS, Dyer SN, Heinrich DA, Bulun SE, Marsh EE, Nowak RA. Reactive oxygen species

mediate mitogenic growth factor signaling pathways in human leiomyoma smooth muscle cells. Biol

Reprod. 2010;82:341-51.

[11] Ono M, Yin P, Navarro A, Moravek MB, Coon JS, Druschitz SA, et al. Paracrine activation of

WNT/β-catenin pathway in uterine leiomyoma stem cells promotes tumor growth. Proc Natl Acad Sci U

S A. 2013;110:17053-8.

[12] Sandberg AA. Updates on the cytogenetics and molecular genetics of bone and soft tissue tumors:

leiomyoma. Cancer Genet Cytogenet. 2005;158:1-26.

[13] Mäkinen N, Mehine M, Tolvanen J, Kaasinen E, Li Y, Lehtonen HJ, et al. MED12, the mediator

complex subunit 12 gene, is mutated at high frequency in uterine leiomyomas. Science. 2011;334:252-5.

[14] Kim M-K, Kim K, Han JY, Lim JM, Song YS. Modulation of inflammatory signaling pathways by

phytochemicals in ovarian cancer. Genes Nutr. 2011;6:109-15.

[15] Vidya Priyadarsini R, Nagini S. Cancer chemoprevention by dietary phytochemicals: promises and

pitfalls. Curr Pharm Biotechnol. 2012;13:125-36.

[16] Surh Y-J. Cancer chemoprevention with dietary phytochemicals. Nat Rev Cancer. 2003;3:768-80.

[17] Chiaffarino F, Parazzini F, La Vecchia C, Chatenoud L, Di Cintio E, Marsico S. Diet and uterine

myomas. Obstet Gynecol. 1999;94:395-8.

[18] Wise LA, Radin RG, Palmer JR, Kumanyika SK, Boggs DA, Rosenberg L. Intake of fruit,

vegetables, and carotenoids in relation to risk of uterine leiomyomata. Am J Clin Nutr. 2011;94:1620-31.

[19] Zhang D, Al-Hendy M, Richard-Davis G, Montgomery-Rice V, Rajaratnam V, Al-Hendy A.

Antiproliferative and proapoptotic effects of epigallocatechin gallate on human leiomyoma cells. Fertil

Steril. 2010;94:1887-93.
27

[20] Zhang D, Al-Hendy M, Richard-Davis G, Montgomery-Rice V, Sharan C, Rajaratnam V, et al.

Green tea extract inhibits proliferation of uterine leiomyoma cells in vitro and in nude mice. Am J Obstet

Gynecol. 2010;202:289. e1-. e9.

[21] Malik M, Mendoza M, Payson M, Catherino WH. Curcumin, a nutritional supplement with

antineoplastic activity, enhances leiomyoma cell apoptosis and decreases fibronectin expression. Fertil

Steril. 2009;91:2177-84.

[22] Catherino WH, Parrott E, Segars J. Proceedings from the National Institute of Child Health and

Human Development Conference on the Uterine Fibroid Research Update Workshop. Fertil Steril.

2011;95:9-12.

[23] Wu C-H, Shieh T-M, Wei L-H, Cheng T-F, Chen H-Y, Huang T-C, et al. Resveratrol inhibits

proliferation of myometrial and leiomyoma cells and decreases extracellular matrix-associated protein

expression. J Funct Foods. 2016;23:241-52.

[24] Kim D, Ramachandran S, Baek S, Kwon SH, Kwon KY, Cha SD, et al. Induction of growth

inhibition and apoptosis in human uterine leiomyoma cells by isoliquiritigenin. Reprod Sci. 2008;15:552-

8.

[25] Moore AB, Castro L, Yu L, Zheng X, Di X, Sifre MI, et al. Stimulatory and inhibitory effects of

genistein on human uterine leiomyoma cell proliferation are influenced by the concentration. Hum

Reprod. 2007;22:2623-31.

[26] Islam MS, Akhtar MM, Ciavattini A, Giannubilo SR, Protic O, Janjusevic M, et al. Use of dietary

phytochemicals to target inflammation, fibrosis, proliferation, and angiogenesis in uterine tissues:

Promising options for prevention and treatment of uterine fibroids? Mol Nutr Food Res. 2014;58:1667-

84.

[27] Di X, Andrews DMK, Tucker CJ, Yu L, Moore AB, Zheng X, et al. A high concentration of

genistein down-regulates activin A, Smad3 and other TGF-β pathway genes in human uterine leiomyoma

cells. Exp Mol Med. 2012;44:281-92.


28

[28] Magalhaes B, Peleteiro B, Lunet N. Dietary patterns and colorectal cancer: systematic review and

meta-analysis. Eur J Cancer Prev. 2012;21:15-23.

[29] Aune D, Chan DSM, Vieira AR, Rosenblatt DAN, Vieira R, Greenwood DC, et al. Fruits, vegetables

and breast cancer risk: a systematic review and meta-analysis of prospective studies. Breast Cancer Res

Treat. 2012;134:479-93.

[30] Paxton RJ, Garcia-Prieto C, Berglund M, Hernandez M, Hajek RA, Handy B, et al. A randomized

parallel-group dietary study for stages II-IV ovarian cancer survivors. Gynecol Oncol. 2012;124:410-6.

[31] Liu RH. Health-promoting components of fruits and vegetables in the diet. Adv Nutr. 2013;4:384S-

92S.

[32] Giampieri F, Forbes-Hernandez TY, Gasparrini M, Alvarez-Suarez JM, Afrin S, Bompadre S, et al.

Strawberry as a health promoter: an evidence based review. Food & function. 2015;6:1386-98.

[33] Liu RH. Dietary bioactive compounds and their health implications. J Food Sci. 2013;78:A18-A25.

[34] Amin ARMR, Kucuk O, Khuri FR, Shin DM. Perspectives for cancer prevention with natural

compounds. J Clin Oncol. 2009;27:2712-25.

[35] Shi Y, Hata A, Lo RS, Massagué J, Pavletich NP. A structural basis for mutational inactivation of the

tumour suppressor Smad4. Nature. 1997;388:87-93.

[36] Itoh F, Asao H, Sugamura K, Heldin CH, ten Dijke P, Itoh S. Promoting bone morphogenetic protein

signaling through negative regulation of inhibitory Smads. EMBO J. 2001;20:4132-42.

[37] Xu J, Luo X, Chegini N. Differential expression, regulation, and induction of Smads, transforming

growth factor-beta signal transduction pathway in leiomyoma, and myometrial smooth muscle cells and

alteration by gonadotropin-releasing hormone analog. J Clin Endocrinol Metab. 2003;88:1350-61.

[38] Heldin CH, Miyazono K, Ten Dijke P. TGF-β signalling from cell membrane to nucleus through

SMAD proteins. Nature. 1997;390:465-71.

[39] Shi Y, Massagué J. Mechanisms of TGF-β signaling from cell membrane to the nucleus. Cell.

2003;113:685-700.
29

[40] Chegini N, Luo X, Ding L, Ripley D. The expression of Smads and transforming growth factor beta

receptors in leiomyoma and myometrium and the effect of gonadotropin releasing hormone analogue

therapy. Mol Cell Endocrinol. 2003;209:9-16.

[41] Arici A, Sozen I. Expression, menstrual cycle-dependent activation, and bimodal mitogenic effect of

transforming growth factor-beta1 in human myometrium and leiomyoma. Am J Obstet Gynecol.

2003;188:76-83.

[42] Luo X, Ding L, Xu J, Chegini N. Gene expression profiling of leiomyoma and myometrial smooth

muscle cells in response to transforming growth factor-beta. Endocrinology. 2005;146:1097-118.

[43] Luo X, Ding L, Chegini N. CCNs, fibulin-1C and S100A4 expression in leiomyoma and

myometrium: inverse association with TGF-beta and regulation by TGF-beta in leiomyoma and

myometrial smooth muscle cells. Mol Hum Reprod. 2006;12:245-56.

[44] Ding L, Xu J, Luo X, Chegini N. Gonadotropin releasing hormone and transforming growth factor

beta activate mitogen-activated protein kinase/extracellularly regulated kinase and differentially regulate

fibronectin, type I collagen, and plasminogen activator inhibitor-1 expression in leiomyoma and

myometrial smooth muscle cells. J Clin Endocrinol Metab. 2004;89:5549-57.

[45] Levens E, Luo X, Ding L, Williams RS, Chegini N. Fibromodulin is expressed in leiomyoma and

myometrium and regulated by gonadotropin-releasing hormone analogue therapy and TGF-beta through

Smad and MAPK-mediated signalling. Mol Hum Reprod. 2005;11:489-94.

[46] Laping NJ, Everitt JI, Frazier KS, Burgert M, Portis MJ, Cadacio C, et al. Tumor-specific efficacy of

transforming growth factor-beta RI inhibition in Eker rats. Clin Cancer Res. 2007;13:3087-99.

[47] Ciarmela P, Carrarelli P, Islam MS, Janjusevic M, Zupi E, Tosti C, et al. Ulipristal acetate modulates

the expression and functions of activin A in leiomyoma cells. Reprod Sci. 2014;21:1120-5.

[48] Engelman JA, Luo J, Cantley LC. The evolution of phosphatidylinositol 3-kinases as regulators of

growth and metabolism. Nat Rev Genet. 2006;7:606-19.


30

[49] Ren Y, Yin H, Tian R, Cui L, Zhu Y, Lin W, et al. Different effects of epidermal growth factor on

smooth muscle cells derived from human myometrium and from leiomyoma. Fertil Steril. 2011;96:1015-

20.

[50] Zhao L, Vogt PK. Class I PI3K in oncogenic cellular transformation. Oncogene. 2008;27:5486-96.

[51] Alessi DR, James SR, Downes CP, Holmes AB, Gaffney PRJ, Reese CB, et al. Characterization of a

3-phosphoinositide-dependent protein kinase which phosphorylates and activates protein kinase Balpha.

Curr Biol. 1997;7:261-9.

[52] Richardson CJ, Schalm SS, Blenis J. PI3-kinase and TOR: PIKTORing cell growth. Semin Cell Dev

Biol. 2004;15:147-59.

[53] Crabtree JS, Jelinsky SA, Harris HA, Choe SE, Cotreau MM, Kimberland ML, et al. Comparison of

human and rat uterine leiomyomata: identification of a dysregulated mammalian target of rapamycin

pathway. Cancer Res. 2009;69:6171-8.

[54] Karra L, Shushan A, Ben-Meir A, Rojansky N, Klein BY, Shveiky D, et al. Changes related to

phosphatidylinositol 3-kinase/Akt signaling in leiomyomas: possible involvement of glycogen synthase

kinase 3alpha and cyclin D2 in the pathophysiology. Fertil Steril. 2010;93:2646-51.

[55] Kovacs KA, Lengyel F, Vértes Z, Környei JL, Gőcze PM, Sumegi B, et al. Phosphorylation of PTEN

(phosphatase and tensin homologue deleted on chromosome ten) protein is enhanced in human

fibromyomatous uteri. J Steroid Biochem Mol Biol. 2007;103:196-9.

[56] Sefton EC, Qiang W, Serna V, Kurita T, Wei J-J, Chakravarti D, et al. MK-2206, an AKT Inhibitor,

promotes caspase-independent cell death and inhibits leiomyoma growth. Endocrinology. 2013;154:4046-

57.

[57] Hoekstra AV, Sefton EC, Berry E, Lu Z, Hardt J, Marsh E, et al. Progestins activate the AKT

pathway in leiomyoma cells and promote survival. J Clin Endocrinol Metab. 2009;94:1768-74.

[58] Boulton TG, Nye SH, Robbins DJ, Ip NY, Radzlejewska E, Morgenbesser SD, et al. ERKs: a family

of protein-serine/threonine kinases that are activated and tyrosine phosphorylated in response to insulin

and NGF. Cell. 1991;65:663-75.


31

[59] Yu L, Saile K, Swartz CD, He H, Zheng X, Kissling GE, et al. Differential expression of receptor

tyrosine kinases (RTKs) and IGF-I pathway activation in human uterine leiomyomas. Mol Med.

2008;14:264-75.

[60] Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell. 2000;103:211-25.

[61] Avruch J, Zhang X-f, Kyriakis JM. Raf meets Ras: completing the framework of a signal

transduction pathway. Trends Biochem Sci. 1994;19:279-83.

[62] Adachi M, Fukuda M, Nishida E. Nuclear export of MAP kinase (ERK) involves a MAP kinase

kinase (MEK)-dependent active transport mechanism. J Cell Biol. 2000;148:849-56.

[63] Yoon S, Seger R. The extracellular signal-regulated kinase: multiple substrates regulate diverse

cellular functions. Growth Factors. 2006;24:21-44.

[64] Shushan A, Rojansky N, Laufer N, Klein BY, Shlomai Z, Levitzki R, et al. The AG1478 tyrosine

kinase inhibitor is an effective suppressor of leiomyoma cell growth. Hum Reprod. 2004;19:1957-67.

[65] Shushan A, Ben-Bassat H, Mishani E, Laufer N, Klein BY, Rojansky N. Inhibition of leiomyoma

cell proliferation in vitro by genistein and the protein tyrosine kinase inhibitor TKS050. Fertil Steril.

2007;87:127-35.

[66] Liang M, Wang H, Zhang Y, Lu S, Wang Z. Expression and functional analysis of platelet-derived

growth factor in uterine leiomyomata. Cancer Biol Ther. 2006;5:28-33.

[67] Gao Z, Matsuo H, Wang Y, Nakago S, Maruo T. Up-regulation by IGF-I of proliferating cell nuclear

antigen and Bcl-2 protein expression in human uterine leiomyoma cells. J Clin Endocrinol Metab.

2001;86:5593-9.

[68] Moon RT, Kohn AD, De Ferrari GV, Kaykas A. WNT and β-catenin signalling: diseases and

therapies. Nat Rev Genet. 2004;5:691-701.

[69] Logan CY, Nusse R. The Wnt signaling pathway in development and disease. Annu Rev Cell Dev

Biol. 2004;20:781-810.

[70] Mikels AJ, Nusse R. Wnts as ligands: processing, secretion and reception. Oncogene. 2006;25:7461-

8.
32

[71] Tanwar PS, Lee H-J, Zhang L, Zukerberg LR, Taketo MM, Rueda BR, et al. Constitutive activation

of Beta-catenin in uterine stroma and smooth muscle leads to the development of mesenchymal tumors in

mice. Biol Reprod. 2009;81:545-52.

[72] Arango NA, Szotek PP, Manganaro TF, Oliva E, Donahoe PK, Teixeira J. Conditional deletion of

beta-catenin in the mesenchyme of the developing mouse uterus results in a switch to adipogenesis in the

myometrium. Dev Biol. 2005;288:276-83.

[73] Markowski DN, Bartnitzke S, Löning T, Drieschner N, Helmke BM, Bullerdiek J. MED12 mutations

in uterine fibroids-their relationship to cytogenetic subgroups. Int J Cancer. 2012;131:1528–36.

[74] Arici A, Sozen I. Transforming growth factor-beta3 is expressed at high levels in leiomyoma where

it stimulates fibronectin expression and cell proliferation. Fertil Steril. 2000;73:1006-11.

[75] Kim S, Xu X, Hecht A, Boyer TG. Mediator is a transducer of Wnt/β-catenin signaling. J Biol Chem.

2006;281:14066-75.

[76] Jin K-S, Oh YN, Hyun SK, Kwon HJ, Kim BW. Betulinic acid isolated from Vitis amurensis root

inhibits 3-isobutyl-1-methylxanthine induced melanogenesis via the regulation of MEK/ERK and

PI3K/Akt pathways in B16F10 cells. Food Chem Toxicol. 2014;68:38-43.

[77] Karna E, Szoka L, Palka JA. Betulinic acid inhibits the expression of hypoxia-inducible factor 1alpha

and vascular endothelial growth factor in human endometrial adenocarcinoma cells. Mol Cell Biochem.

2010;340:15-20.

[78] Viji V, Shobha B, Kavitha SK, Ratheesh M, Kripa K, Helen A. Betulinic acid isolated from Bacopa

monniera (L.) Wettst suppresses lipopolysaccharide stimulated interleukin-6 production through

modulation of nuclear factor-B in peripheral blood mononuclear cells. Int Immunopharmacol.

2010;10:843-9.

[79] Szuster-Ciesielska A, Plewka K, Daniluk J, Kandefer-Szerszeń M. Betulin and betulinic acid

attenuate ethanol-induced liver stellate cell activation by inhibiting reactive oxygen species (ROS),

cytokine (TNF-α, TGF-β) production and by influencing intracellular signaling. Toxicology.

2011;280:152-63.
33

[80] Khan N, Adhami VM, Afaq F, Mukhtar H. Butein induces apoptosis and inhibits prostate tumor

growth in vitro and in vivo. Antioxid Redox Signal. 2012;16:1195-204.

[81] Cho SG, Woo SM, Ko SG. Butein suppresses breast cancer growth by reducing a production of

intracellular reactive oxygen species. J Exp Clin Cancer Res. 2014;33:51.

[82] Zhang L, Chen W, Li X. A novel anticancer effect of butein: Inhibition of invasion through the

ERK1/2 and NF-B signaling pathways in bladder cancer cells. FEBS Lett. 2008;582:1821-8.

[83] Chung C-H, Chang C-H, Chen S-S, Wang H-H, Yen J-Y, Hsiao C-J, et al. Butein inhibits

angiogenesis of human endothelial progenitor cells via the translation dependent signaling pathway. Evid

Based Complement Alternat Med. 2013;2013:943187.

[84] Moon D-O, Choi YH, Moon S-K, Kim W-J, Kim G-Y. Butein suppresses the expression of nuclear

factor-kappa B-mediated matrix metalloproteinase-9 and vascular endothelial growth factor in prostate

cancer cells. Toxicol In Vitro. 2010;24:1927-34.

[85] Jang JH, Yang ES, Min K-J, Kwon TK. Inhibitory effect of butein on tumor necrosis factor-α-

induced expression of cell adhesion molecules in human lung epithelial cells via inhibition of reactive

oxygen species generation, NF-κB activation and Akt phosphorylation. Int J Mol Med. 2012;30:1357-64.

[86] Lee SD, Choe JW, Lee BJ, Kang MH, Joo MK, Kim JH, et al. Butein effects in colitis and

interleukin-6/signal transducer and activator of transcription 3 expression. World Journal of

Gastroenterology: WJG. 2015;21:465-74.

[87] Wang Z, Lee Y, Eun JS, Bae EJ. Inhibition of adipocyte inflammation and macrophage chemotaxis

by butein. Eur J Pharmacol. 2014;738:40-8.

[88] Szuster-Ciesielska A, Mizerska-Dudka M, Daniluk J, Kandefer-Szerszen„ M. Butein inhibits

ethanol-induced activation of liver stellate cells through TGF-β, NFκB, p38, and JNK signaling pathways

and inhibition of oxidative stress. J Gastroenterol. 2013;48:222-37.

[89] Park S-Y, Kim J-Y, Lee S-M, Jun C-H, Cho S-B, Park C-H, et al. Capsaicin induces apoptosis and

modulates MAPK signaling in human gastric cancer cells. Mol Med Rep. 2014;9:499-502.
34

[90] Lee S-H, Richardson RL, Dashwood RH, Baek SJ. Capsaicin represses transcriptional activity of β-

catenin in human colorectal cancer cells. J Nutr Biochem. 2012;23:646-55.

[91] Min J-K, Han K-Y, Kim E-C, Kim Y-M, Lee S-W, Kim O-H, et al. Capsaicin inhibits in vitro and in

vivo angiogenesis. Cancer Res. 2004;64:644-51.

[92] Bhutani M, Pathak AK, Nair AS, Kunnumakkara AB, Guha S, Sethi G, et al. Capsaicin is a novel

blocker of constitutive and interleukin-6 -inducible STAT3 activation. Clin Cancer Res. 2007;13:3024-

32.

[93] Chakraborty S, Adhikary A, Mazumdar M, Mukherjee S, Bhattacharjee P, Guha D, et al. Capsaicin-

induced activation of p53-SMAR1 auto-regulatory loop down-regulates VEGF in non-small cell lung

cancer to restrain angiogenesis. PloS one. 2014;9:e99743.

[94] Bitencourt S, de Mesquita FC, Caberlon E, da Silva GV, Basso BS, Ferreira GA, et al. Capsaicin

induces de-differentiation of activated hepatic stellate cell. Biochem Cell Biol. 2012;90:683-90.

[95] Park J-Y, Kawada T, Han I-S, Kim B-S, Goto T, Takahashi N, et al. Capsaicin inhibits the

production of tumor necrosis factor α by LPS-stimulated murine macrophages, RAW 264.7: a PPARγ

ligand-like action as a novel mechanism. FEBS Lett. 2004;572:266-70.

[96] Kim C-S, Kawada T, Kim B-S, Han I-S, Choe S-Y, Kurata T, et al. Capsaicin exhibits anti-

inflammatory property by inhibiting IkB-a degradation in LPS-stimulated peritoneal macrophages. Cell

Signal. 2003;15:299-306.

[97] Lim W, Jeong W, Song G. Delphinidin suppresses proliferation and migration of human ovarian

clear cell carcinoma cells through blocking AKT and ERK1/2 MAPK signaling pathways. Mol Cell

Endocrinol. 2016;422:172-81.

[98] Pal HC, Sharma S, Strickland LR, Agarwal J, Athar M, Elmets CA, et al. Delphinidin reduces cell

proliferation and induces apoptosis of non-small-cell lung cancer cells by targeting EGFR/VEGFR2

signaling pathways. PloS one. 2013;8:e77270.

[99] Keravis Trs, Favot L, Abusnina AA, Anton A, Justiniano Hln, Soleti R, et al. Delphinidin Inhibits

Tumor Growth by Acting on VEGF Signalling in Endothelial Cells. PloS one. 2015;10:e0145291.
35

[100] Duluc L, Jacques C, Soleti R, Andriantsitohaina R, Simard G. Delphinidin inhibits VEGF induced-

mitochondrial biogenesis and Akt activation in endothelial cells. Int J Biochem Cell Biol. 2014;53:9-14.

[101] Lamy S, Beaulieu E, Labbe D, Bedard V, Moghrabi A, Barrette S, et al. Delphinidin, a dietary

anthocyanidin, inhibits platelet-derived growth factor ligand/receptor (PDGF/PDGFR) signaling.

Carcinogenesis. 2008;29:1033-41.

[102] Lim T-G, Jung SK, Kim J-e, Kim Y, Lee HJ, Jang TS, et al. NADPH oxidase is a novel target of

delphinidin for the inhibition of UVB-induced MMP-1 expression in human dermal fibroblasts. Exp

Dermatol. 2013;22:428-30.

[103] Cho J-S, Kang J-H, Shin J-M, Park I-H, Lee H-M. Inhibitory effect of delphinidin on extracellular

matrix production via the MAPK/NF-κB pathway in nasal polyp-derived fibroblasts. Allergy Asthma

Immunol Res. 2015;7:276-82.

[104] Weng JR, Bai LY, Chiu CF, Wang YC, Tsai MH. The dietary phytochemical 3,3'-diindolylmethane

induces G2/M arrest and apoptosis in oral squamous cell carcinoma by modulating Akt-NF-B, MAPK,

and p53 signaling. Chem Biol Interact. 2012;195:224-30.

[105] Rahman KW, Sarkar FH. Inhibition of nuclear translocation of nuclear factor-kB contributes to 3,3'-

diindolylmethane-induced apoptosis in breast cancer cells. Cancer Res. 2005;65:364-71.

[106] Zhu J, Li Y, Guan C, Chen Z. Anti-proliferative and pro-apoptotic effects of 3,3'-diindolylmethane

in human cervical cancer cells. Oncol Rep. 2012;28:1063-8.

[107] Leem S-H, Li XJ, Park MH, Park BH, Kim SM. Genome-wide transcriptome analysis reveals

inactivation of Wnt/β-catenin by 3,3'-diindolylmethane inhibiting proliferation of colon cancer cells. Int J

Oncol. 2015;47:918-26.

[108] Kandala PK, Wright SE, Srivastava SK. Blocking epidermal growth factor receptor activation by 3,

3′-diindolylmethane suppresses ovarian tumor growth in vitro and in vivo. J Pharmacol Exp Ther.

2012;341:24-32.

[109] Chang X, Firestone GL, Bjeldanes LF. Inhibition of growth factor-induced Ras signaling in

vascular endothelial cells and angiogenesis by 3,3'-diindolylmethane. Carcinogenesis. 2006;27:541-50.


36

[110] Kunimasa K, Kobayashi T, Kaji K, Ohta T. Antiangiogenic effects of indole-3-carbinol and 3,3'-

diindolylmethane are associated with their differential regulation of ERK1/2 and Akt in tube-forming

HUVEC. J Nutr. 2010;140:1-6.

[111] Kong D, Banerjee S, Huang W, Li Y, Wang Z, Kim H-RC, et al. Mammalian target of rapamycin

repression by 3,3'-diindolylmethane inhibits invasion and angiogenesis in platelet-derived growth factor-

D-overexpressing PC3 cells. Cancer Res. 2008;68:1927-34.

[112] Kim HW, Kim J, Kim J, Lee S, Choi B-R, Han J-S, et al. 3, 3′-Diindolylmethane inhibits

lipopolysaccharide-induced microglial hyperactivation and attenuates brain inflammation. Toxicol Sci.

2014;137:158-67.

[113] Zhang Z, Gao Z, Hu W, Yin S, Wang C, Zang Y, et al. 3, 3' Diindolylmethane ameliorates

experimental hepatic fibrosis via inhibiting miR-21 expression. Br J Pharmacol. 2013;170:649-60.

[114] Li J, Zhang W, Jiao R, Yang Z, Yuan Y, Wu Q, et al. DIM attenuates TGF-β1-induced

myofibroblast differentiation in neonatal rat cardiac fibroblasts. Int J Clin Exp Pathol. 2015;8:5121-8.

[115] Subramaniam A, Shanmugam MK, Ong TH, Li F, Perumal E, Chen L, et al. Emodin inhibits

growth and induces apoptosis in an orthotopic hepatocellular carcinoma model by blocking activation of

STAT3. Br J Pharmacol. 2013;170:807-21.

[116] Pooja T, Karunagaran D. Emodin suppresses Wnt signaling in human colorectal cancer cells

SW480 and SW620. Eur J Pharmacol. 2014;742:55-64.

[117] Way T-D, Huang J-T, Chou C-H, Huang C-H, Yang M-H, Ho C-T. Emodin represses TWIST1-

induced epithelial-mesenchymal transitions in head and neck squamous cell carcinoma cells by inhibiting

the beta-catenin and Akt pathways. Eur J Cancer. 2014;50:366-78.

[118] Thacker PC, Karunagaran D. Curcumin and emodin down-regulate TGF-β signaling pathway in

human cervical cancer cells. PloS one. 2015;10:e0120045.

[119] Kaneshiro T, Morioka T, Inamine M, Kinjo T, Arakaki J, Chiba I, et al. Anthraquinone derivative

emodin inhibits tumor-associated angiogenesis through inhibition of extracellular signal-regulated kinase

1/2 phosphorylation. Eur J Pharmacol. 2006;553:46-53.


37

[120] Pang X, Liu J, Li Y, Zhao J, Zhang X. Emodin inhibitshomocysteine-induced C-reactive protein

generation in vascular smooth muscle cells by regulating PPARγ expression and ROS-ERK1/2/p38 signal

pathway. PloS one. 2015;10:e0131295.

[121] Yin X, Gong X, Jiang R, Kuang G, Wang B, Zhang L, et al. Emodin ameliorated

lipopolysaccharide-induced fulminant hepatic failure by blockade of TLR4/MD2 complex expression in

D-galactosamine-sensitized mice. Int Immunopharmacol. 2014;23:66-72.

[122] Li D, Zhang N, Cao Y, Zhang W, Su G, Sun Y, et al. Emodin ameliorates lipopolysaccharide-

induced mastitis in mice by inhibiting activation of NF-κB and MAPKs signal pathways. Eur J

Pharmacol. 2013;705:79-85.

[123] Lee J, Jung E, Lee J, Huh S, Hwang C-H, Lee H-Y, et al. Emodin inhibits TNF alpha-induced

MMP-1 expression through suppression of activator protein-1 (AP-1). Life Sci. 2006;79:2480-5.

[124] Wang R, Wan Q, Zhang Y, Huang F, Yu K, Xu D, et al. Emodin suppresses interleukin-1 beta

induced mesangial cells proliferation and extracellular matrix production via inhibiting P38 MAPK. Life

Sci. 2007;80:2481-8.

[125] Li X, Liu W, Wang Q, Liu P, Deng Y, Lan T, et al. Emodin suppresses cell proliferation and

fibronectin expression via p38MAPK pathway in rat mesangial cells cultured under high glucose. Mol

Cell Endocrinol. 2009;307:157-62.

[126] Ambothi K, Prasad NR, Balupillai A. Ferulic acid inhibits UVB-radiation induced

photocarcinogenesis through modulating inflammatory and apoptotic signaling in Swiss albino mice.

Food Chem Toxicol. 2015;82:72-8.

[127] Yang G-W, Jiang J-S, Lu W-Q. Ferulic acid exerts anti-angiogenic and anti-tumor activity by

targeting fibroblast growth factor receptor 1-mediated angiogenesis. Int J Mol Sci. 2015;16:24011-31.

[128] Hou Y, Yang J, Zhao G, Yuan Y. Ferulic acid Inhibits endothelial cell proliferation through NO

down-regulating ERK1/2 pathway. J Cell Biochem. 2004;93:1203-9.

[129] Xu T, Pan Z, Dong M, Yu C, Niu Y. Ferulic acid suppresses activation of hepatic stellate cells

through ERK1/2 and Smad signaling pathways in vitro. Biochem Pharmacol. 2015;93:49-58.
38

[130] Wei M-g, Sun W, He W-m, Ni L, Yang Y-y. Ferulic acid attenuates TGF-β1-induced renal cellular

fibrosis in NRK-52E cells by inhibiting Smad/ILK/Snail pathway. Evid Based Complement Alternat

Med. 2015;2015:619720.

[131] Kim H-Y, Lee S-M. Ferulic acid attenuates ischemia/reperfusion-induced hepatocyte apoptosis via

inhibition of JNK activation. Eur J Pharm Sci. 2012;45:708-15.

[132] Khan N, Afaq F, Khusro FH, Mustafa AV, Suh Y, Mukhtar H. Dual inhibition of

phosphatidylinositol 3-kinase/Akt and mammalian target of rapamycin signaling in human nonsmall cell

lung cancer cells by a dietary flavonoid fisetin. Int J Cancer. 2012;130:1695-705.

[133] Suh Y, Afaq F, Khan N, Johnson JJ, Khusro FH, Mukhtar H. Fisetin induces autophagic cell death

through suppression of mTOR signaling pathway in prostate cancer cells. Carcinogenesis. 2010;31:1424-

33.

[134] Zhang X-J, Jia S-S. Fisetin inhibits laryngeal carcinoma through regulation of AKT/NF-κB/mTOR

and ERK1/2 signaling pathways. Biomed Pharmacother. 2016;83:1164-74.

[135] Bhat TA, Nambiar D, Pal A, Agarwal R, Singh RP. Fisetin inhibits various attributes of

angiogenesis in vitro and in vivo--implications for angioprevention. Carcinogenesis. 2011;33:385-93.

[136] Park H-H, Lee S, Oh J-M, Lee M-S, Yoon K-H, Park BH, et al. Anti-inflammatory activity of

fisetin in human mast cells (HMC-1). Pharmacol Res. 2007;55:31-7.

[137] Kim S-C, Kang S-H, Jeong S-J, Kim S-H, Ko HS, Kim S-H. Inhibition of c-Jun N-terminal kinase

and nuclear factor κB pathways mediates fisetin-exerted anti-inflammatory activity in lipopolysccharide-

treated RAW264.7 cells. Immunopharmacol Immunotoxicol. 2012;34:645-50.

[138] Lee J-D, Huh J-E, Jeon G, Yang H-R, Woo H-S, Choi D-Y, et al. Flavonol-rich RVHxR from Rhus

verniciflua Stokes and its major compound fisetin inhibits inflammation-related cytokines and angiogenic

factor in rheumatoid arthritic fibroblast-like synovial cells and in vivo models. Int Immunopharmacol.

2009;9:268-76.
39

[139] Kim S-H, Hwang K-A, Choi K-C. Treatment with kaempferol suppressed breast cancer cell growth

caused by estrogen and triclosan in cellular and xenograft breast cancer models. J Nutr Biochem.

2016;28:70-82.

[140] Yao S, Wang X, Li C, Zhao T, Jin H, Fang W. Kaempferol inhibits cell proliferation and glycolysis

in esophagus squamous cell carcinoma via targeting EGFR signaling pathway. Tumor Biology.

2016;37:10247-56.

[141] Dang Q, Song W, Xu D, Ma Y, Li F, Zeng J, et al. Kaempferol suppresses bladder cancer tumor

growth by inhibiting cell proliferation and inducing apoptosis. Mol Carcinog. 2015;54:831-40.

[142] Kim S-Y, Jin Y-R, Lim Y, Kim J-H, Cho M-R, Hong J-T, et al. Inhibition of PDGF beta-receptor

tyrosine phosphorylation and its downstream intracellular signal transduction in rat aortic vascular smooth

muscle cells by kaempferol. Planta Med. 2005;71:599-603.

[143] Lee HS, Cho HJ, Kwon GT, Park JHY. Kaempferol downregulates insulin-like growth factor-I

receptor and ErbB3 signaling in HT-29 human colon cancer cells. J Cancer Prev. 2014;19:161–9.

[144] Luo H, Rankin GO, Liu L, Daddysman MK, Jiang B-H, Chen YC. Kaempferol inhibits

angiogenesis and VEGF expression through both HIF dependent and independent pathways in human

ovarian cancer cells. Nutr Cancer. 2009;61:554-63.

[145] Mylonis I, Lakka A, Tsakalof A, Simos G. The dietary flavonoid kaempferol effectively inhibits

HIF-1 activity and hepatoma cancer cell viability under hypoxic conditions. Biochem Biophys Res

Commun. 2010;398:74-8.

[146] Luo H, Rankin GO, Juliano N, Jiang B-H, Chen YC. Kaempferol inhibits VEGF expression and in

vitro angiogenesis through a novel ERK-NFκB-cMyc-p21 pathway. Food Chem. 2012;130:321-8.

[147] Chen X, Yang X, Liu T, Guan M, Feng X, Dong W, et al. Kaempferol regulates MAPKs and NF-

κB signaling pathways to attenuate LPS-induced acute lung injury in mice. Int Immunopharmacol.

2012;14:209-16.
40

[148] Suchal K, Malik S, Gamad N, Malhotra RK, Goyal SN, Chaudhary U, et al. Kaempferol attenuates

myocardial ischemic injury via inhibition of MAPK signaling pathway in experimental model of

myocardial ischemia-reperfusion injury. Oxid Med Cell Longev. 2016;2016:7580731.

[149] Yoon H-Y, Lee E-G, Lee H, Cho IJ, Choi YJ, Sung M-S, et al. Kaempferol inhibits IL-1β-induced

proliferation of rheumatoid arthritis synovial fibroblasts and the production of COX-2, PGE2 and MMPs.

Int J Mol Med. 2013;32:971-7.

[150] Gong J-H, Cho I-H, Shin D, Han S-Y, Park S-H, Kang Y-H. Inhibition of airway epithelial-to-

mesenchymal transition and fibrosis by kaempferol in endotoxin-induced epithelial cells and ovalbumin-

sensitized mice. Lab Invest. 2013;94:297-308.

[151] Jin H, Lee WS, Eun SY, Jung JH, Park H-S, Kim G, et al. Morin, a flavonoid from Moraceae,

suppresses growth and invasion of the highly metastatic breast cancer cell line MDA-MB-231 partly

through suppression of the Akt pathway. Int J Oncol. 2014;45:1629-37.

[152] Ma Y, Ge A, Zhu W, Liu Y-N, Ji N-F, Zha W-J, et al. Morin attenuates ovalbumin-induced airway

inflammation by modulating oxidative stress-responsive MAPK signaling. Oxid Med Cell Longev.

2016;2016:5843672.

[153] Dhanasekar C, Kalaiselvan S, Rasool M. Morin, a bioflavonoid suppresses monosodium urate

crystal-induced inflammatory immune response in RAW 264.7 macrophages through the inhibition of

inflammatory mediators, intracellular ROS levels and NF-B activation. PloS one. 2015;10:e0145093.

[154] MadanKumar P, NaveenKumar P, Manikandan S, Devaraj H, NiranjaliDevaraj S. Morin

ameliorates chemically induced liver fibrosis in vivo and inhibits stellate cell proliferation in vitro by

suppressing Wnt/β-catenin signaling. Toxicol Appl Pharmacol. 2014;277:210-20.

[155] Li H, Yang B, Huang J, Xiang T, Yin X, Wan J, et al. Naringin inhibits growth potential of human

triple-negative breast cancer cells by targeting β-catenin signaling pathway. Toxicol Lett. 2013;220:219-

28.
41

[156] Raha S, Yumnam S, Hong GE, Lee HJ, Saralamma VVG, Park H-S, et al. Naringin induces

autophagy-mediated growth inhibition by downregulating the PI3K/Akt/mTOR cascade via activation of

MAPK pathways in AGS cancer cells. Int J Oncol. 2015;47:1061-9.

[157] Zhang Y-S, Li Y, Wang Y, Sun S-Y, Jiang T, Li C, et al. Naringin, a natural dietary compound,

prevents intestinal tumorigenesis in Apc Min/+ mouse model. J Cancer Res Clin Oncol. 2016;142:913-25.

[158] Zeng L, Zhen Y, Chen Y, Zou L, Zhang Y, Hu F, et al. Naringin inhibits growth and induces

apoptosis by a mechanism dependent on reduced activation of NF-B/COX-2-caspase-1 pathway in HeLa

cervical cancer cells. Int J Oncol. 2014;45:1929-36.

[159] Lee E-J, Kim D-I, Kim W-J, Moon S-K. Naringin inhibits matrix metalloproteinase-9 expression

and AKT phosphorylation in tumor necrosis factor-alpha-induced vascular smooth muscle cells. Mol Nutr

Food Res. 2009;53:1582-91.

[160] Chen Y, Nie Y-C, Luo Y-L, Lin F, Zheng Y-F, Cheng G-H, et al. Protective effects of naringin

against paraquat-induced acute lung injury and pulmonary fibrosis in mice. Food Chem Toxicol.

2013;58:133–40.

[161] Chen F, Zhang N, Ma X, Huang T, Shao Y, Wu C, et al. Naringin alleviates diabetic kidney disease

through inhibiting oxidative stress and inflammatory reaction. PloS one. 2015;10:e0143868.

[162] Tsai M-L, Lai C-S, Chang Y-H, Chen W-J, Ho C-T, Pan M-H. Pterostilbene, a natural analogue of

resveratrol, potently inhibits 7, 12-dimethylbenz [a] anthracene (DMBA)/12-O-tetradecanoylphorbol-13-

acetate (TPA)-induced mouse skin carcinogenesis. Food & function. 2012;3:1185-94.

[163] Paul S, Rimando AM, Lee HJ, Ji Y, Reddy BS, Suh N. Anti-inflammatory action of pterostilbene is

mediated through the p38 mitogen-activated protein kinase pathway in colon cancer cells. Cancer Prev

Res (Phila). 2009;2:650-7.

[164] Paul S, DeCastro AJ, Lee HJ, Smolarek AK, So JY, Simi B, et al. Dietary intake of pterostilbene, a

constituent of blueberries, inhibits the β-catenin/p65 downstream signaling pathway and colon

carcinogenesis in rats. Carcinogenesis. 2010;31:1272-8.


42

[165] Chiou Y-S, Tsai M-L, Wang Y-J, Cheng A-C, Lai W-M, Badmaev V, et al. Pterostilbene inhibits

colorectal aberrant crypt foci (ACF) and colon carcinogenesis via suppression of multiple signal

transduction pathways in azoxymethane-treated mice. J Agric Food Chem. 2010;58:8833-41.

[166] Pan M-H, Chiou Y-S, Chen W-J, Wang J-M, Badmaev V, Ho C-T. Pterostilbene inhibited tumor

invasion via suppressing multiple signal transduction pathways in human hepatocellular carcinoma cells.

Carcinogenesis. 2009;30:1234-42.

[167] Zhang Y, Zhang Y. Pterostilbene, a novel natural plant conduct, inhibits high fat-induced

atherosclerosis inflammation via NF-κB signaling pathway in Toll-like receptor 5 (TLR5) deficient mice.

Biomed Pharmacother. 2016;81:345-55.

[168] Li J, Deng R, Hua X, Zhang L, Lu F, Coursey TG, et al. Blueberry component pterostilbene

protects corneal epithelial cells from inflammation via anti-oxidative pathway. Sci Rep. 2016;6:19408.

[169] Lee MF, Liu ML, Cheng AC, Tsai ML, Ho CT, Liou WS, et al. Pterostilbene inhibits

dimethylnitrosamine-induced liver fibrosis in rats. Food Chem. 2013;138:802–7.

[170] Singh RP, Gu M, Agarwal R. Silibinin inhibits colorectal cancer growth by inhibiting tumor cell

proliferation and angiogenesis. Cancer Res. 2008;68:2043-50.

[171] Ravichandran K, Velmurugan B, Gu M, Singh RP, Agarwal R. Inhibitory effect of silibinin against

azoxymethane-induced colon tumorigenesis in A/J mice. Clin Cancer Res. 2010;16:4595-606.

[172] Raina K, Agarwal C, Agarwal R. Effect of silibinin in human colorectal cancer cells: Targeting the

activation of NF-κB signaling. Mol Carcinog. 2013;52:195-206.

[173] Kim S, Han J, Kim JS, Kim J-H, Choe J-H, Yang J-H, et al. Silibinin suppresses EGFR ligand-

induced CD44 expression through inhibition of EGFR activity in breast cancer cells. Anticancer Res.

2011;31:3767-73.

[174] Lee M-H, Huang Z, Kim DJ, Kim S-H, Kim MO, Lee S-Y, et al. Direct targeting of MEK1/2 and

RSK2 by silybin induces cell-cycle arrest and inhibits melanoma cell growth. Cancer Prev Res (Phila).

2013;6:455-65.
43

[175] Lin CH, Li CH, Liao PL, Tse LS, Huang WK, Cheng HW, et al. Silibinin inhibits VEGF secretion

and age-related macular degeneration in a hypoxia-dependent manner through the PI-3 kinase/Akt/mTOR

pathway. Br J Pharmacol. 2013;168:920-31.

[176] Garcia-Maceira P, Mateo J. Silibinin inhibits hypoxia-inducible factor-1alpha and

mTOR/p70S6K/4E-BP1 signalling pathway in human cervical and hepatoma cancer cells: implications

for anticancer therapy. Oncogene. 2009;28:313-24.

[177] Park J-W, Shin N-R, Shin I-S, Kwon O-K, Kim J-S, Oh S-R, et al. Silibinin inhibits neutrophilic

inflammation and mucus secretion induced by cigarette smoke via suppression of ERK‐SP1 Pathway.

Phytother Res. 2016: [Epub ahead of print].

[178] Choi YH, Jin GY, Guo HS, Piao HM, chang Li L, Li GZ, et al. Silibinin attenuates allergic airway

inflammation in mice. Biochem Biophys Res Commun. 2012;427:450-5.

[179] Liu W, Li Y, Zheng X, Zhang K, Du Z. Potent inhibitory effect of silibinin from milk thistle on skin

inflammation stimuli by 12-O-tetradecanoylphorbol-13-acetate. Food & function. 2015;6:3712-9.

[180] Ai W, Zhang Y, Tang QZ, Yan L, Bian ZY, Liu C, et al. Silibinin attenuates cardiac hypertrophy

and fibrosis through blocking EGFR-dependent signaling. J Cell Biochem. 2010;110:1111-22.

[181] Chen YH, Liang CM, Chen CL, Chen JT, Chang YH, Lu DW, et al. Silibinin inhibits myofibroblast

transdifferentiation in human tenon fibroblasts and reduces fibrosis in a rabbit trabeculectomy model.

Acta Ophthalmol (Copenh). 2013;97:e506–e15.

[182] Cho JW, Il KJ, Lee KS. Downregulation of type I collagen expression in silibinin-treated human

skin fibroblasts by blocking the activation of Smad2/3-dependent signaling pathways: Potential

therapeutic use in the chemoprevention of keloids. Int J Mol Med. 2013;31:1148-52.

[183] Trappoliere M, Caligiuri A, Schmid M, Bertolani C, Failli P, Vizzutti F, et al. Silybin, a component

of sylimarin, exerts anti-inflammatory and anti-fibrogenic effects on human hepatic stellate cells. J

Hepatol. 2009;50:1102-11.
44

[184] Rajput S, Kumar BNP, Dey KK, Pal I, Parekh A, Mandal M. Molecular targeting of Akt by

thymoquinone promotes G 1 arrest through translation inhibition of cyclin D1 and induces apoptosis in

breast cancer cells. Life Sci. 2013;93:783-90.

[185] Sutton KM, Greenshields AL, Hoskin DW. Thymoquinone, a bioactive component of black

caraway seeds, causes G1 phase cell cycle arrest and apoptosis in triple-negative breast cancer cells with

mutant p53. Nutr Cancer. 2014;66:408-18.

[186] Xu D, Ma Y, Zhao B, Li S, Zhang Y, Pan S, et al. Thymoquinone induces G2/M arrest, inactivates

PI3K/Akt and nuclear factorκB pathways in human cholangiocarcinomas both in vitro and in vivo. Oncol

Rep. 2014;31:2063-70.

[187] Yang J, Kuang X-r, Lv P-t, Yan X-x. Thymoquinone inhibits proliferation and invasion of human

nonsmall-cell lung cancer cells via ERK pathway. Tumour Biol. 2015;36:259-69.

[188] Yi T, Cho S-G, Yi Z, Pang X, Rodriguez M, Wang Y, et al. Thymoquinone inhibits tumor

angiogenesis and tumor growth through suppressing AKT and extracellular signal-regulated kinase

signaling pathways. Mol Cancer Ther. 2008;7:1789-96.

[189] Peng L, Liu A, Shen Y, Xu H-Z, Yang S-Z, Ying X-Z, et al. Antitumor and anti-angiogenesis

effects of thymoquinone on osteosarcoma through the NF-B pathway. Oncol Rep. 2013;29:571-8.

[190] Su X, Ren Y, Yu N, Kong L, Kang J. Thymoquinone inhibits inflammation, neoangiogenesis and

vascular remodeling in asthma mice. Int Immunopharmacol. 2016;38:70-80.

[191] Wang D, Qiao J, Zhao X, Chen T, Guan D. Thymoquinone Inhibits IL-1β-Induced Inflammation in

Human Osteoarthritis Chondrocytes by Suppressing NF-κB and MAPKs Signaling Pathway.

Inflammation. 2015;38:2235-41.

[192] Umar S, Hedaya O, Singh AK, Ahmed S. Thymoquinone inhibits TNF-α-induced inflammation and

cell adhesion in rheumatoid arthritis synovial fibroblasts by ASK1 regulation. Toxicol Appl Pharmacol.

2015;287:299-305.
45

[193] Bai T, Yang Y, Wu Y-L, Jiang S, Lee JJ, Lian L-H, et al. Thymoquinone alleviates thioacetamide-

induced hepatic fibrosis and inflammation by activating LKB1-AMPK signaling pathway in mice. Int

Immunopharmacol. 2014;19:351-7.

[194] Bai T, Lian L-H, Wu Y-L, Wan Y, Nan J-X. Thymoquinone attenuates liver fibrosis via PI3K and

TLR4 signaling pathways in activated hepatic stellate cells. Int Immunopharmacol. 2013;15:275-81.

[195] Ghazwani M, Zhang Y, Gao X, Fan J, Li J, Li S. Anti-fibrotic effect of thymoquinone on hepatic

stellate cells. Phytomedicine. 2013;21:254-60.

[196] Pourgholamhossein F, Sharififar F, Rasooli R, Pourgholi L, Nakhaeipour F, Samareh-Fekri H, et al.

Thymoquinone effectively alleviates lung fibrosis induced by paraquat herbicide through down-regulation

of pro-fibrotic genes and inhibition of oxidative stress. Environ Toxicol Pharmacol. 2016;45:340-5.

[197] El-Khouly D, El-Bakly WM, Awad A, El-Mesalamy H, El-Demerdash E. Thymoquinone blocks

lung injury and fibrosis by attenuating bleomycin-induced oxidative stress and activation of nuclear factor

Kappa-B in rats. Toxicology. 2012; 302 106–13.


46

Figure legends

Fig. 1. Dietary phytochemicals and their chemical structure and dietary sources.

Fig. 2A-B. Smad 2/3 (A) and PI3K (B) signaling pathways in uterine leiomyoma targeted by dietary

phytochemicals.

Fig. 3A-B. ERK 2/3 (A) and β-catenin (B) signaling pathways in uterine leiomyoma targeted by dietary

phytochemicals.
47

Figure 1
48

Figure 2
49

Figure 3

You might also like