You are on page 1of 11

doi: 10.1111/jop.

12171

J Oral Pathol Med (2015) 44: 167177


2014 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
wileyonlinelibrary.com/journal/jop

Molecular and cellular cues of diet-associated oral


carcinogenesiswith an emphasis on areca-nut-induced
oral cancer development
Wan-Chun Li1,2,3, Pei-Lun Lee2, I-Chiang Chou1,4, Wan-Jung Chang2, Shu-Chun Lin1,2,5, Kuo-Wei
Chang1,2,5
1

Department of Dentistry, School of Dentistry, National Yang-Ming University, Taipei, Taiwan; 2Institute of Oral Biology, School of
Dentistry, National Yang-Ming University, Taipei, Taiwan; 3Department of Education and Research, Taipei City Hospital, Taipei, Taiwan;
4
Department of Dentistry, Zhong-Xiao Branch, Taipei City Hospital, Taipei, Taiwan; 5Department of Stomatology, Taipei Veterans
General Hospital, Taipei, Taiwan

In modern times, potent dietary carcinogens are key


contributors for neoplastic development. For oral squamous cell carcinoma (OSCC), one of the leading cancer
types in developing countries, main oncogenic inducers/
enhancers, including areca nut chewing, tobacco smoking, and alcohol consumption, were shown to promote
cancer initiation/progression. Over decades, studies from
different laboratories have identified underlying cellular
and molecular mechanisms for carcinogen-induced
OSCC. In this review, we will give an overview of where
we are in understanding potential oral carcinogenic
factors stimulated OSCC tumorigenesis, especially those
associated with areca nut chewing in Asians, aiming to
provide future scope of possible interception.
J Oral Pathol Med (2015) 44: 167177
Keywords: areca nut; curcumin; oral cancer; tea polyphenol

Introduction
The formation of oral squamous cell carcinoma (OSCC), a
subcategory of head and neck cancers, is the outcome of
multiple hit-and-go events acquired mostly from oral intake,
which affects oral epithelial/mucosal tissues. Overall intraand extracellular carcinogenic input triggers aberrant genetic
accumulation, defective epigenetic regulation, and pathological microenvironment resulting in sequential pathological
transformation (1). The neoplastic process could be evident
by several abnormal pathological features (hyperkeratosis,
epithelial hyperplasia, and dysplasia), phenotypic alterations
Correspondence: Kuo-Wei Chang, DDS, PhD, Department of Dentistry,
School of Dentistry, National Yang-Ming University, No. 155, Li-Nong St.,
Sec.2, Taipei, Taiwan 112. Tel: +8862-28267223, Fax: +8862-28264053,
E-mail: ckcw@ym.edu.tw
Accepted for publication January 23, 2014

(invasion, enhanced chemoattraction of lymphocytes, and


angiogenesis) as well as leukoplakia, erythroleukoplakia,
lichenoid changes, and other pre-cancerous lesions clinically
identied (2). Several dened oral carcinogens or tumorpromoting materials, including alcohol consumption, tobacco
smoking, areca nut chewing, and viral infection, are risk
factors for oral malignancy (36). At the molecular level,
earlier studies were initiated by examination of differential
gene mutations or aberrant expression in cancer cells/tissues
compared with normal oral tissues. Many studies from our
laboratory showed that deregulated expression of genes for
proliferation, cell mobility, survival, angiogenesis, and
immune modulation could be implicated for areca-associated
OSCC pathogenesis (714). The importance of environmental challenges including infection of oncogenic human
papillomavirus (e.g., HPV16, 18, 31) (1517) and imbalanced metabolism such as diabetes mellitus (1822) linking
to oral carcinogenesis has also been discussed elsewhere. In
contrast, various anti-oncogenic natural compounds such as
tea polyphenols and curcumin were also found to counteract
oral carcinogenesis by means of various regulations (23, 24).
To further explore mechanisms underlying different
stages of oral tumorigenesis, tissues from normal, pre-cancerous, early, and late stages of tumors are required to
elucidate dynamic pathological process of oral carcinogenesis. However, the use of clinical samples only represented
snapshots but not systemic conditions of disease
progression, while the limited access to clinical samples
from the same individuals restricted comprehensive
interpretation of cellular and molecular cues important for
oral cancer formation. Most mechanistic analysis was
therefore performed in in vitro cultured oral cells. Despite
the treatment in vitro may not truly reect in vivo
metabolism of candidate oncogenic substances, both acute
and prolonged cytotoxic response in cultured cells, either
from isolated primary oral epithelial/mesenchymal cells or
established OSCC cell lines, are indeed of great help to
identify essential mechanisms of OSCC formation. Rodent

Overview of diet-mediated oral cancer


Li et al.

168

models were also applied to induce oral neoplastic lesions in


vivo by treatment for various dietary carcinogens using
different methods including painting onto buccal pouch or
administration in drinking water (2527). More recently,
identication of epigenetic modulators such as non-coding
small RNAs for OSCC pathology was also carried out.
For example, disruption was frequently identied in OSCC
(2831). Taken together, this review aims to provide an
overview of carcinogenic effects of dietary uptakes, with
main emphasis on areca nut constitutes.

Areca (betel) nut chewing vs. oral


carcinogenesis
Areca nut is a nut of tropic palm. Areca quid (or called betel
quid in some regions) contains areca nut, inorescence piper
betel (also called betel leaf), slaked lime, tobacco, liqueed
catechu, and other spicy ingredients (32). Areca chewing is a
popular oral habit in South Asia; it is the fourth most common
addictive substance. Despite the different ways in which
areca nut is consumed in various regions, areca chewing
manifests general pharmacological effects, including euphoria, central nervous system stimulation, vertigo, salivation,
miosis, tremor, and bradycardia. Physiologically, areca
chewing could produce a sense of well-being, euphoria,
heightened alertness, sweating, salivation, and increased
body temperature leading to habituation and addiction,
mainly via the modulation on the central and autonomic
nervous system (33). Epidemiological evidences showed that
areca use is closely associated with various oral pathogenesis
such as oral submucous brosis (OSF), OSCC, periodontal
disorders, and others (34). Areca-associated OSCC is the
third most common neoplasia in developing countries,
comprising around 50% of all malignancies in some nations
of South Asia (3537). An early study demonstrated
epidermal thickening of the tongue, esophagus, and forestomach of rats in groups fed with areca nut diet for 480 days,
implying its carcinogenic activity (38). Although malignant
tumors were not activated in this experiment, the observation
of pre-cancerous-like epithelial lesions suggested other
milder carcinogens or cocarcinogens in areca nut. It was
reported that areca chewers often swallow the juice in direct
contact with oropharynx and esophagus, thereby leading to a
higher risk of cancer in these anatomic sites, indicating that
the quid also contains other milder carcinogens or cocarcinogens, possibly mediated by the formation of areca-nutspecic or tobacco-specic N-nitrosamines (39, 40). In
addition, Atkinson et al. (41) reported that a high incidence
of OSCC in the New Guinea population might be resulted
from the habit of chewing areca quid, even though the areca
quid which is never mixed with tobacco, implying that areca
nut could also have a carcinogenic effect when chewed
without tobacco (42).
Areca nut itself comprises a number of compounds
identied as potential tumor inducers or enhancers. For
example, alkaloid is one of the main constituents in areca
nut. Around 8595% of total alkaloid content in areca nut is
arecoline, while others are arecaidine, guvacoline, and
guvacine (43). In addition to areca nut alkaloids, studies
have been carried out to test the mutagenic and carcinogenic
effects of other alternative constituents in areca quid using
J Oral Pathol Med

different experimental models. These components include


polyphenols, tannin, metal ions, and safrole. Studies have
identied areca ingredients as the synergistic or promoting
agents for multistep chemical carcinogenesis in different
animal models (8, 44). Nonetheless, the extracted areca nut
compounds described in previous sections to monitor the
physiological impact on oral tumorigenesis are pure chemicals, raising an argument whether the effect of the treatment
could truly recapitulate physiological conditions of oral cells
exposed to areca nut consumption environment. To address
this issue, the crude extract from areca nut (ANE) was
chosen to use in many studies. ANE could be prepared in
two ways, based on the involvement of areca nut husk,
thereby resulting in ripe ANE (rANE, used in most studies)
and tender ANE (tANE) for research uses (45, 46). The
extract of areca nut is tumorigenic on mouse skin and elicits
sarcoma in the connective tissue of mice and rats. It also
induces hepatocellular carcinoma (HCC) in mice upon
gastric intubation, suggesting that ANE is capable of
stimulating cancer formation (47). As ANE-induced oral
cancer progression was emphasized, the established OSCC
cell lines were often used. Interestingly, the molecular clues
underlying ANE-mediated tumor development are similar to
the factors involved in cancer initiation, although the
administrative concentration of ANE (approximately 5 lg/
ml) in normal human oral keratinocyte (NHOK)/normal
human oral broblast (NHOF) is much lower than the dose
treated in OSCC cell lines (approximately 20 lg/ml).
Areca quid-induced cytotoxicity and genotoxicity
Early studies indicated that arecoline treatment led to
increased sister chromatid exchange in mouse bone marrow
cells as well as higher frequency of micronuclei, an
unscheduled mitotic debris during cell division, in Chinese
hamster ovary cells (48, 49). Further investigation suggested
that arecoline could induce cell cycle arrest at prometaphase
by deregulating mitotic spindle assembly supporting the role
of arecoline in producing progeny that carried various
chromosomal abnormalities and genomic instability (50). It
was shown that the expression of p53-regulated p21 and
p53-activated DNA repair was repressed by arecoline
administration in human epithelial cells, suggesting that
arecoline-mediated p53 inhibition plays an important role in
areca-nut-associated tumorigenesis (51). As for the effect on
primary oral cells, arecoline was cytotoxic but not genotoxic
to cultured human buccal broblasts, and this arecolinemediated cytotoxicity could be overcome after the treatment
of antioxidants glutathione and glycyrrhizin (52). The
synergistic effect with the cigarette smoking product
nicotine was found to enhance arecoline-associated cytotoxicity (53). To explore susceptible genes accounting for
arecoline-induced cellular damage, a high-throughput
genetic screening for human gingival broblasts treated
with or without arecoline was carried out. Twelve candidate
genes associated with xenobiotic metabolism, maintenance
of genomic stability, DNA damage or repair, stress
response, and the TGFb signaling pathway were uncovered,
presenting potential targets for how arecoline induced
cytotoxicity and initiated cellular impairments (54).
Using in vivo models, the tumorigenic effect of areca nut
alkaloids was also investigated. Lin et al. (55) demonstrated

Overview of diet-mediated oral cancer


Li et al.

172

Areca nut
extract

Tobacco
ingredients
Tumor progression:
- ERK
Cox2
PGE2
Vimen n
- Akt
Vimen n
Cell mo lity
- NF-B
[ROS]
p38
HIF1
Autophagy
Curcumin
Tea polyphenols

Overdosed
Alcohol

Tumor ini a on:


- Genotoxicity
- NF-B
ROS
Cytotoxicity

MMP2

Areca nut
extract

Niche eect:
- Invasion
- Akt
Focal adhesion

Normal oral epithelium

Neoplas c oral
epithelium

ANE s mulated oral


fibroblasts

Premalignant oral
epithelium

Normal oral fibroblasts

Cancer associated
fibroblasts

Figure 1 Diagraphic illustration of dietary-associated oncogenic effects. The tumorigenic promoters including areca nut, tobacco, and alcohol could trigger
various cellular and molecular events key for oral cancer formation, while curcumin and tea polyphenols suppress cancerous phenotypes by antagonistic
mechanisms.

4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone and N-nitrosonornicotine, polycyclic aromatic hydrocarbons (e.g.,


benzo[a]pyrene), and aromatic amines have the most potent
links to OSCC, mainly through induction of DNA adducts
(117). Ethanol itself is not carcinogenic in animal study, as its
metabolites such as acetaldehyde could cause DNA damage
leading to higher risk of OSCC. It is interesting to note that
the anatomic sites in close contact with alcohol are at higher
risk to develop tumors, indicating the facilitating role of
alcohol for oral tumor formation (118). Epidemiologically, a
recent large cohort analysis conducted by the International
Head and Neck Cancer Epidemiology (INHANCE) consortium showed the attributable risk of smoking and drinking
combined for cancer formation to be 64%, implying the joint
effect of these two components in major proportion of
patients with head and neck cancer (119). Bruguere et al.
(120) found relative risk value of 13.5 for OSCC when 100
159 g of alcohol was consumed daily, while men who both
smoke and drink are nearly 38 times more likely to develop
head and neck cancers than men who do neither (121). A
casecontrol study showed that the odds ratio was 123 times
for Taiwanese patients with OSCC who smoked, drank
alcohol, and chewed areca quid than non-users, suggesting
synergetic effect of alcohol and tobacco consumption in
areca nut chewers (122). The underlying molecular cues for
smoking and drinking are still poorly understood. Early
studies using PCR-single-strand conformation polymorphism and DNA sequencing to analyze the conserved exon
regions (exon 59) of p53 gene in OSCC specimens
identied that tobacco smoking drives an important contribution to p53 mutation, while alcohol might enhance these
mutagenic effects (109). Overall, alcohol consumption and
smoking could be dened as enhancers for areca-associated
oral carcinogenesis, although further analysis for molecular
players for this synergetic impact would be required.
J Oral Pathol Med

Anti-oncogenic dietary moleculestea


polyphenols and curcumin
From the aspect of chemoprevention, the use of natural
substances derived from diet could provide a safe strategy to
inhibit cancer with limited toxicity. Two such agents, green
tea and the spice curcumin, have been extensively investigated (123126). In general, tea polyphenols and curcumin
could inhibit oral malignancy via a number of cellular and
molecular mechanismsmost of those targets shared signals involved in areca-associated oral oncology (127129).
Recent studies determined that curcumin hinders arecoline
or ANE-associated oral cancer development, probably via
the regulation of heat-shock protein, HIF, or placenta
growth factor (130132).
Several studies identied the benets of using tea
polyphenols for suppression of cancer development, mainly
through its major constituent ()-epigallocatechin-3-gallate
(EGCG) (133). Using cultured OSCC cells, in vitro effects
of EGCG contained several key steps: (1) to inhibit cell
proliferation via ROS-mediated apoptosis induction and cell
cycle arrest as well as telomerase activity; (2) to modulate
transcription factors such as NF-jB and activator protein-1
(AP1); and (3) to reduce cell migration and invasion by
decreased expression of MMPs and urokinase plasminogen
activator serine protease (u-PA) (134136). As for animal
models of oral carcinogenesis, treatment with 0.6% green
tea reduced the number of visible tumors by 35% and
reduced tumor volume by 57% in the DMBA-induced
hamster buccal pouch cancers. Immunohistochemical analyses showed that tea increased the apoptotic index of the
tumors as well as decreased the proliferation index and
microvessel density (137). Recent examination giving green
tea polyphenols extracts to 4-nitroquinoline 1-oxide (4NQO) induced oral tongue carcinogenesis in rats was

Overview of diet-mediated oral cancer


Li et al.

170

Table 2 Oncogenic effects of areca nut extract treatment


Cell type
Mouse skin/liver
Human normal
oral epithelium

Human OSCC
cells

Human normal oral


broblasts

Effects
Sarcoma/hepatocellular
carcinoma
Increased senescence
Hyperdiploid chromosomal
changes
Cell cycle arrest
Increased PI3K/Akt ?
Increased dedifferentiation
Increased Rac/Rho/
Phospho-JNK
Increased broblastoids
Increased stress ber/
lamellipodin
Increased NF-kB ? Increased
Cox2/ProstaglandinE2/IL6/TNFa
Increased JNK/ERK/p38 ?
Increased inammation
Increased MMP9 ? Increased
NFkB ? Decreased cell
viability/Decreased re-epithelization
Increased lysyl oxidase activity
Muscarinic M4 receptor-mediated
signaling ? Increased cell motility
Increased oxidative stress
? Increased
HIF1a ? Increased autophagy
Increased NF-kB ? Increased
miR146a
Increased AuroA ? Increased
autophagy
Increased senescence
Cell cycle arrest
Increased MMP2 secretion ?
Increased PBMC invasiveness
Decreased FANCG ? Decreased
DNA repair ? Increased miR23a
Increased Proto-oncogene c-fos/c-jun

References
47
6668

74
80, 82

110,111
112
113
12
81
102
106
107
73
73, 84
105
108

(76). Further study to examine the effect of arecoline on the


expression of Cyr61, an important matricellular protein in
promoting many types of tumors, revealed that arecoline
stimulated Cyr61 synthesis in human gingival epithelial SG
cells leading to putative carcinogenesis. The Cyr61-mediated tumorigenic effect could be attenuated with the
treatment of various inhibitors to abrogate ERK, reactive
oxidative species (ROS), ROCK, and HMG-CoA reductase,
demonstrating distinctive molecular regulations may underlie arecoline-related oral pathogenesis (77). The regulation
of environmental-sensing integrins upon areca nut ingredient treatment was also reported. The up-regulation of
integrin avb6 was detected in approximately 50% of human
OSF suggesting integrin may be a potential component
involving in arecoline-induced oral aberrations. A recent
study using oral keratinocyte-derived cells genetically
modied to express high avb6 indeed showed that arecoline-dependent avb6 up-regulation promoted keratinocyte
migration and induced invasion, providing a possibility that
this mechanism may support malignant transformation (78).
In addition to areca nut alkaloids, safrole treatment could
also regulate tissue inhibitor of metalloproteinase-1 (TIMP1), a key matrix remodeling protein in isolated buccal
mucosal broblasts from patients with OSF, providing a
possible pathologic mechanism of OSF (79).
J Oral Pathol Med

The treatment with ANE induced the transition of


polygonal morphology into broblastoid, formation of
lamellipodia, and stress bers in NHOK and OECM-1 cells,
which indicated that ANE administration impaired cytoskeletal network of oral keratinocytes. Further mechanistic
analysis showed that active Rac and Rho GTPase and
phosphorylation of JNK could be important in regulating
morphological changes under ANE treatment (80). A recent
study deciphered that the stimulation of a muscarinic M4
receptor-mediated signaling cascade by ANE could also
promote the migration of OSCC cells. The increased cell
motility was regulated via the activation of Src and ERK
kinases (81). In both clinical and cellular model, investigations showed the association of areca-stimulated vimentin
expression, cell mobility, and progression of OSCC,
implying tumorigenic effect of ANE could be through
regulation of cytoskeletal proteins (82). Moreover, elevated
content of metal ion copper in the areca nut was detected in
OSF tissues, supporting the idea that copper is an initiating
factor for OSF while this pathological brogenic stimulation
may be mediated by the up-regulation of lysyl oxidase
(LOX) activity (83). The contribution of this metastaticrelated enzyme LOX using both in vitro and in vivo oral
cancer pathogenesis was further proved in a later study.
LOX was enhanced upon ANE treatment through AKT
activation. As the LOX expression was associated with cell
migration and invasion, and tumorigenesis, this indicated
that LOX facilitated oncogenic transformation in OSCC
cells (12). Taking the niche effect into account, the ANEactivated conditional medium of NHOF promoted various
malignant properties of OSCC cells, probably via the
paracrine activity of MMP-2 produced from NHOF (73). In
addition, the MMP-2 secreted by ANE-treated NHOF also
enhanced the invasiveness of polymorphonuclear leukocytes, indicating the relevance between inammatory processes with oral carcinogenesis in areca chewers (84).
Epigenetic manipulation and microRNA changes in arecanut-associated oral carcinogenesis
The molecular cues underlying the change in cellular
phenotype or gene expression other than the change in
gene sequences could be dened as epigenetic regulations.
A recent report implied that another class of regulatory
player of areca-nut-associated pathogenesis is via
epigenetic modulation. Using histological analysis, the
frequency of hypermethylation on Ras association domaincontaining protein 1A (RASSF1A) and p16 promoter in
OSCC and verrucous carcinoma (VC) tissues (93% and
63% in OSCC and 100% and 67% in VC for RASSF1A
and p16, respectively) was reported recently (85). Upon the
exposure of arecoline, human leukemia K-562 cells
exhibited epigenetic alterations based on the detection of
attenuation of a panel of genes catalyzing histone methylation, acetylation, and demethylation (86). In addition,
OSCC-related microRNAs (miRNAs) were also identied.
Once discovered, miRNAs have drawn lots of attention in
medical research not only because of their role in
regulating various physiological conditions but also due
to great potential to serve as biomarkers for early diagnosis
and prognosis of diseases (87, 88). miRNA is a class
of small single-stranded RNAs comprised of 22- to

Overview of diet-mediated oral cancer


Li et al.

25-nucleotide-long non-coding RNA that normally bind to


the 30 untranslated region (30 UTR) of their target mRNA,
leading to translational inhibition and/or mRNA degradation (89). The miRNAs are initially transcribed as long
primary miRNAs, which are processed to a hairpin
structure in the nucleus by RNase Drosha, and further
cleaved to miRNA duplexes by RNase Dicer in cytosol.
The miRNA duplexes are then unwound, and one strand,
termed mature miRNA or guide strand, which has
complimentarity to target mRNAs, is loaded into the
RNA-induced silencing complex. The complimentarity of
miRNAs to target RNAs renders the cleavage or the
translational repression of targeted mRNA (9092). Over
500 miRNAs have been discovered in human genome, and
it has been estimated that they could regulate 7492% of
all protein encoding mRNAs to a certain extent (93).
Considering the complex level of gene expression regulation conferred by miRNAs, miRNAs could regulate a wide
range of biological events including cell growth and
proliferation, differentiation, organogenesis, metabolism,
stress response, stemness, tissue remodeling, and onset/
progression of different diseases such as cancer, cardiovascular diseases, and diabetes through epigenetic regulation (9496).
In general, oncogenic miRNAs are usually overexpressed, driving tumor initiation and progression; tumor
suppressive miRNAs are down-regulated or deleted facilitating cancer development (97). The miRNA expression
proles appear to be tumor- and tissue specic (98). As for
OSCC development, many miRNAs play important roles in
regulating cell malignancy (99, 100). The dysregulation of
these cancerous miRNAs in OSCC induces cell proliferation and anti-apoptosis, promotes cancer metastasis, and
mediates resistance to chemotherapy (94). Among them,
OSCC-related oncogenic miRNAs included miR-21, miR31, miR-184, and miR-221, while let-7, miR-1, miR-133a,
miR-138, miR-145, and miR-375 have shown to be tumor
suppressive (101). The activation of the hypoxia pathway is
important for the pathogenesis of OSCC. ANE up-regulates
hypoxia-inducible factor 1a (HIF1a) in OSCC cells (102).
MiR-31 was identied to be important for OSCC progression by impeding factor-inhibiting hypoxia-inducible factor
to activate HIF based on different in vitro and in vivo assays
(103). Further study showed that miR-210 also targeted
hypoxic markers, including HIF-1a, TWIST, and carbonic
anhydrase IX (CA IX), to correlate tumor recurrence and
reduced survival rate (104). Very recently, studies also
revealed that that ANE could trigger aberrant miRNA
expression. For instance, ANE-induced miR-23a expression
correlated with increased DNA damage proteins and
down-regulated DNA double-strand breaks repair machinery. This ANE-stimulated miR-23a was mediated with a
reduced expression of Fanconis anemia susceptibility genes
(FANCG) that participate in a double-strand break repair
pathway to prevent chromosomal aberrations (105).
Furthermore, ANE treatment up-regulates miR-146a
expression, likely through the activation of NF-jB (106).
Overall, further examination would be required to elucidate
the ANE-induced miRNA expression prole for better
identication of the role of diet-induced miRNA alteration
in oral carcinogenesis.

Areca-nut-induced autophagy
Autophagy is a catabolic mechanism in which cells degrade
unnecessary organelles to ensure survival in adverse
environments. In the regards to long-term incubation of
areca consumption in vivo, a more recent study further
demonstrated that low-dose ANE treatment in OSCC cells
induced oxidative stress and up-regulated HIF1a, and this
therefore led to autophagy (102). The results showed that
10 lg/ml ANE administration on SAS OSCC cells resulted
in acidic vesicles evidenced by acridine orange staining
analysis, LC3-II accumulation, and autophagosomes
genesis. While the blockage of ROS, HIF1a induction,
p38-mediated signal activity, and other events decreased
ANE-induced autophagy. Taken together, the incubation of
ANE in OSCC cells exhibited a protective effect to decrease
cell apoptosis, while ANE treatment induced various
mitogen-mediated signaling pathways, leading to the promotion of cell malignancy. Interestingly, knockdown of
Aurora A oncogene in OSCC cells also resulted in the
autophagy, implicating that this inhibition might be
important for oral cancer therapy (107).

171

Other molecular cues underlying areca-mediated


oncogenesis
Numbers of studies have sought to explore detailed cellular
and molecular events after treatment of areca-nut-associated
constitutes. The effect of ANE treatment on primary
cultured oral cells was initially observed by the detection
of increased proto-oncogenes c-fos and c-jun in NHOF
(108). The correlation between p53 mutation and the
presence of safrole-DNA adducts in oral cancer patients
with areca-nut-chewing history supporting that safrole could
be one of the key players causing p53 mutation (109). Other
studies revealed that ANE could induce an inammatory
response based on the detection of increased production of
COX-2, prostaglandin E2, interleukin 6, and tumor necrosis
factor a (110, 111), while a later study showed that ANEmediated COX-2 overexpression was probably evoked
through induction of mitogen-activated protein kinases
(e.g., JNK1, ERK, and p38) and NF-jB activation (112).
NF-jB activation was shown upon ANE treatment in arecaassociated OC3 cells (67), while similar results were
reported in non-areca-associated OSCC cell lines (112). A
more recent study linking the increased inammation,
reduced differentiation, and altered cell structural protein
organization to ANE. It showed that ANE could activate
NF-jB activation for MMP-9 up-regulation to decrease cell
viability and impede re-epithelization of normal oral
keratinocyte (113). The schematic diagram in Figure 1
summarizes the molecular elements that are known to be
involved in the neoplastic process of oral epithelium, which
are modulated by areca ingredients.

Non-areca-nut-associated oral carcinogenesis


In addition to areca nut, smoking and alcohol consumption
are two other main oncogenic risk factors for oral
tumor development (114, 115). The IARC identied more
than 60 carcinogens in cigarette smoke, and at least 16
carcinogens in unburned tobacco have also been
reported (116). The tobacco-specic nitrosamines including
J Oral Pathol Med

Overview of diet-mediated oral cancer


Li et al.

172

Areca nut
extract

Tobacco
ingredients
Tumor progression:
- ERK
Cox2
PGE2
Vimen n
- Akt
Vimen n
Cell mo lity
- NF-B
[ROS]
p38
HIF1
Autophagy
Curcumin
Tea polyphenols

Overdosed
Alcohol

Tumor ini a on:


- Genotoxicity
- NF-B
ROS
Cytotoxicity

MMP2

Areca nut
extract

Niche eect:
- Invasion
- Akt
Focal adhesion

Normal oral epithelium

Neoplas c oral
epithelium

ANE s mulated oral


fibroblasts

Premalignant oral
epithelium

Normal oral fibroblasts

Cancer associated
fibroblasts

Figure 1 Diagraphic illustration of dietary-associated oncogenic effects. The tumorigenic promoters including areca nut, tobacco, and alcohol could trigger
various cellular and molecular events key for oral cancer formation, while curcumin and tea polyphenols suppress cancerous phenotypes by antagonistic
mechanisms.

4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone and N-nitrosonornicotine, polycyclic aromatic hydrocarbons (e.g.,


benzo[a]pyrene), and aromatic amines have the most potent
links to OSCC, mainly through induction of DNA adducts
(117). Ethanol itself is not carcinogenic in animal study, as its
metabolites such as acetaldehyde could cause DNA damage
leading to higher risk of OSCC. It is interesting to note that
the anatomic sites in close contact with alcohol are at higher
risk to develop tumors, indicating the facilitating role of
alcohol for oral tumor formation (118). Epidemiologically, a
recent large cohort analysis conducted by the International
Head and Neck Cancer Epidemiology (INHANCE) consortium showed the attributable risk of smoking and drinking
combined for cancer formation to be 64%, implying the joint
effect of these two components in major proportion of
patients with head and neck cancer (119). Bruguere et al.
(120) found relative risk value of 13.5 for OSCC when 100
159 g of alcohol was consumed daily, while men who both
smoke and drink are nearly 38 times more likely to develop
head and neck cancers than men who do neither (121). A
casecontrol study showed that the odds ratio was 123 times
for Taiwanese patients with OSCC who smoked, drank
alcohol, and chewed areca quid than non-users, suggesting
synergetic effect of alcohol and tobacco consumption in
areca nut chewers (122). The underlying molecular cues for
smoking and drinking are still poorly understood. Early
studies using PCR-single-strand conformation polymorphism and DNA sequencing to analyze the conserved exon
regions (exon 59) of p53 gene in OSCC specimens
identied that tobacco smoking drives an important contribution to p53 mutation, while alcohol might enhance these
mutagenic effects (109). Overall, alcohol consumption and
smoking could be dened as enhancers for areca-associated
oral carcinogenesis, although further analysis for molecular
players for this synergetic impact would be required.
J Oral Pathol Med

Anti-oncogenic dietary moleculestea


polyphenols and curcumin
From the aspect of chemoprevention, the use of natural
substances derived from diet could provide a safe strategy to
inhibit cancer with limited toxicity. Two such agents, green
tea and the spice curcumin, have been extensively investigated (123126). In general, tea polyphenols and curcumin
could inhibit oral malignancy via a number of cellular and
molecular mechanismsmost of those targets shared signals involved in areca-associated oral oncology (127129).
Recent studies determined that curcumin hinders arecoline
or ANE-associated oral cancer development, probably via
the regulation of heat-shock protein, HIF, or placenta
growth factor (130132).
Several studies identied the benets of using tea
polyphenols for suppression of cancer development, mainly
through its major constituent ()-epigallocatechin-3-gallate
(EGCG) (133). Using cultured OSCC cells, in vitro effects
of EGCG contained several key steps: (1) to inhibit cell
proliferation via ROS-mediated apoptosis induction and cell
cycle arrest as well as telomerase activity; (2) to modulate
transcription factors such as NF-jB and activator protein-1
(AP1); and (3) to reduce cell migration and invasion by
decreased expression of MMPs and urokinase plasminogen
activator serine protease (u-PA) (134136). As for animal
models of oral carcinogenesis, treatment with 0.6% green
tea reduced the number of visible tumors by 35% and
reduced tumor volume by 57% in the DMBA-induced
hamster buccal pouch cancers. Immunohistochemical analyses showed that tea increased the apoptotic index of the
tumors as well as decreased the proliferation index and
microvessel density (137). Recent examination giving green
tea polyphenols extracts to 4-nitroquinoline 1-oxide (4NQO) induced oral tongue carcinogenesis in rats was

Overview of diet-mediated oral cancer


Li et al.

carried out. The decreased average number and volume of


tumors were detected in test groups compared with control
rats (138). Despite all experimental results supporting the
anticancer effect of using tea polyphenols for oral carcinogenesis, different clinical trials showed inconsistent outcomes, indicating the potential discrepancies between
human and rodents, or the needs to select more suitable
subset of subjects for clinical trial (139, 140).
Curcumin is one of the major phenolic antioxidant and
anti-inammatory agents in the spice turmeric (Curcuma
longa). There are three major curcuminoids that constitute
curcumin: curcumin (curcumin I, 75%), demethoxycurcumin (curcumin II, 20%), and bisdemethoxycurcumin
(curcumin III, 5%) (141). Early studies have demonstrated
the activity of curcumin to repress different cancer growth,
including skin and gastrointestinal tract using animal
models (142, 143). The mechanistic analysis indicated
that curcumin had versatile potential to suppress cancer
growth by down-regulation of proto-oncogene expression
and protein kinase activity, suppressed inammation,
inhibition of angiogenesis, and DNA-mutation-mediated
genetic instability (144). Various cell culture studies have
shown that curcumin induced apoptosis in malignant cells
by inhibition of different transcription factors and secondary messengers such as NF-jB, AP1, c-Jun, and JakSTAT signal (145, 146). In addition, curcumin targeted
elements in the mTOR pathway for cancer abrogation
(147). Curcumin also increased the levels of several
endogenous antioxidants via the Nrf2 pathway to
strengthen self-defense mechanism against ROS (148).
More recent investigations demonstrated that curcumin
regulated ANE-associated tumorigenic phenotypes, including autophagy, cell mobility, IGFBP-5-mediated mitogenic
signaling activity, and broblast-tumor cell interaction,
leading to decreased OSCC development (23, 113, 149,
150). Curcumin is also proven for its potential to attenuate
carcinogenesis induced by chemical carcinogens, at both
initiation and progression stages under various preclinical
studies (151). In addition, a recent study identied that
curcumin increased miR-145 expression and decreased
Sox9-mediated regulation cascade, which underlie the
elimination of stemness in OSCC (152). However, there
is an urgent need to validate the efcacy of curcumin in
adjuvant therapy or in prevention of oral carcinogenesis.

Conclusions and future perspective


Oral neoplastic formation is the cellular process in response
to numbers of aberrant genetic and epigenetic regulations
resulting from environmental challenges. Among all oncogenic stimuli, areca nut ingredients play an essential role in
regulating oncogenic phenotypes. It could be concluded that
areca nut induced cytotoxicity and genotoxicity in normal
oral epithelium leading to greater susceptibility for tumor
formation. Prolonged incubation of areca nut ingredients
provides substantial oncogenic inputs resulting in cellular
transformation of normal oral epithelium into malignant
cells, mainly by the activation of various intrinsic tumorigenic signals, including ERK, AKT, and NF-jB pathways.
Interestingly, the external oncogenic contributions from
surrounding stroma for cancer progression have been also

recognized. In contrast, the studies showed that curcumin


and tea polyphenols play antagonistic roles to abrogate oral
carcinogenesis by targeting areca-nut-related molecular cues
(Figure 1).
Although much effort has been made to uncover detailed
molecular cues of diet-mediated carcinogenesis, how to
dene better experimental models in order to truly recapitulate physiological pharmacokinetics of these diet compounds on either normal or malignant oral cells remains a
big challenge. Moreover, the limited success of chemopreventive natural compounds in the clinical setting due
probably to their poor bioavailability, rapid rate of metabolism, or both, restrict the application in medical practice
would also need to be addressed. A new strategy for
development of optimal delivery systems to bypass poor
oral bioavailability as well as to reduce hepatic metabolism
to reach an effective therapeutic concentration in the blood
stream would be a key in the future to improve the success
of OSCC interception.

173

References
1. Da Silva SD, Ferlito A, Takes RP, et al. Advances and
applications of oral cancer basic research. Oral Oncol 2011;
47: 78391.
2. Lambert R, Sauvaget C, De Camargo Cancela M, Sankaranarayanan R. Epidemiology of cancer from the oral cavity
and oropharynx. Eur J Gastroenterol Hepatol 2011; 23:
63341.
3. Amarasinghe HK, Usgodaarachchi US, Johnson NW, Lalloo
R, Warnakulasuriya S. Betel-quid chewing with or without
tobacco is a major risk factor for oral potentially malignant
disorders in Sri Lanka: a case-control study. Oral Oncol
2010; 46: 297301.
4. Reichart PA, Mohr U, Srisuwan S, Geerlings H, Theetranont
C, Kangwanpong T. Precancerous and other oral mucosal
lesions related to chewing, smoking and drinking habits in
Thailand. Community Dent Oral Epidemiol 1987; 15: 15260.
5. Subapriya R, Thangavelu A, Mathavan B, Ramachandran
CR, Nagini S. Assessment of risk factors for oral squamous
cell carcinoma in Chidambaram, Southern India: a casecontrol study. Eur J Cancer Prev 2007; 16: 2516.
6. Yang YY, Koh LW, Tsai JH, et al. Involvement of viral and
chemical factors with oral cancer in Taiwan. Jpn J Clin
Oncol 2004; 34: 17683.
7. Chiang WC, Wong YK, Lin SC, Chang KW, Liu CJ.
Increase of MMP-13 expression in multi-stage oral carcinogenesis and epigallocatechin-3-gallate suppress MMP-13
expression. Oral Dis 2006; 12: 2733.
8. Lin SC, Chang KW, Chang CS, Yu SY, Chao SY, Wong
YK. Establishment and characterization of a cell line
(HCDB-1) derived from a hamster buccal pouch carcinoma
induced by DMBA and Taiwanese betel quid extract. Proc
Natl Sci Counc Repub China B 2000; 24: 12935.
9. Lin SC, Wang CP, Chen YM, et al. Regulation of IGFBP-5
expression during tumourigenesis and differentiation of oral
keratinocytes. J Pathol 2002; 198: 31725.
10. Lingen MW, Chang KW, Mcmurray SJ, et al. Overexpression of p53 in squamous cell carcinoma of the tongue in
young patients with no known risk factors is not associated
with mutations in exons 59. Head Neck 2000; 22: 32835.
11. Liu CJ, Chang KW, Chao SY, et al. The molecular markers
for prognostic evaluation of areca-associated buccal squamous cell carcinoma. J Oral Pathol Med 2004; 33: 32734.

J Oral Pathol Med

Overview of diet-mediated oral cancer


Li et al.

174

12. Shieh TM, Lin SC, Liu CJ, Chang SS, Ku TH, Chang KW.
Association of expression aberrances and genetic polymorphisms of lysyl oxidase with areca-associated oral tumorigenesis. Clin Cancer Res 2007; 13: 437885.
13. Wong YK, Lin SC, Chang CS, et al. Cyclin D1 genotype in
areca-associated oral squamous cell carcinoma. J Oral
Pathol Med 2003; 32: 26570.
14. Wu HT, Ko SY, Fong JH, Chang KW, Liu TY, Kao SY.
Expression of phosphorylated Akt in oral carcinogenesis and
its induction by nicotine and alkaline stimulation. J Oral
Pathol Med 2009; 38: 20613.
15. Hillbertz NS, Hirsch JM, Jalouli J, Jalouli MM, Sand L.
Viral and molecular aspects of oral cancer. Anticancer Res
2012; 32: 420112.
16. Ororke MA, Ellison MV, Murray LJ, Moran M, James J,
Anderson LA. Human papillomavirus related head and neck
cancer survival: a systematic review and meta-analysis. Oral
Oncol 2012; 48: 1191201.
17. Schiffman M, Clifford G, Buonaguro FM. Classication of
weakly carcinogenic human papillomavirus types: addressing the limits of epidemiology at the borderline. Infect Agent
Cancer 2009; 4: 8.
18. Albrecht M, Banoczy J, Dinya E, Tamas G Jr. Occurrence of
oral leukoplakia and lichen planus in diabetes mellitus.
J Oral Pathol Med 1992; 21: 3646.
19. Dikshit RP, Ramadas K, Hashibe M, Thomas G, Somanathan T, Sankaranarayanan R. Association between diabetes
mellitus and pre-malignant oral diseases: a cross sectional
study in Kerala, India. Int J Cancer 2006; 118: 4537.
20. Tseng CH. Oral cancer in Taiwan: is diabetes a risk factor?
Clin Oral Invest 2013; 17: 135764.
21. Vairaktaris E, Goutzanis L, Kalokerinos G, et al. Increased
risk of oral cancer in diabetic animals is not associated with
c-jun activation pathway. J Craniomaxillofac Surg 2007; 35:
3827.
22. Wu CH, Wu TY, Li CC, Lui MT, Chang KW, Kao SY.
Impact of diabetes mellitus on the prognosis of patients with
oral squamous cell carcinoma: a retrospective cohort study.
Ann Surg Oncol 2010; 17: 217583.
23. Chang KW, Hung PS, Lin IY, et al. Curcumin upregulates
insulin-like growth factor binding protein-5 (IGFBP-5) and
C/EBPalpha during oral cancer suppression. Int J Cancer
2010; 127: 920.
24. Narotzki B, Levy Y, Aizenbud D, Reznick AZ. Green tea
and its major polyphenol EGCG increase the activity of oral
peroxidases. Adv Exp Med Biol 2013; 756: 99104.
25. Chen YK, Lin LM. DMBA-induced hamster buccal pouch
carcinoma and VX2-induced rabbit cancer as a model for
human oral carcinogenesis. Expert Rev Anticancer Ther
2010; 10: 148596.
26. Kanojia D, Vaidya MM. 4-nitroquinoline-1-oxide induced
experimental oral carcinogenesis. Oral Oncol 2006; 42:
65567.
27. Vairaktaris E, Spyridonidou S, Papakosta V, et al. The
hamster model of sequential oral oncogenesis. Oral Oncol
2008; 44: 31524.
28. Lin SC, Liu CJ, Lin JA, Chiang WF, Hung PS, Chang KW.
miR-24 up-regulation in oral carcinoma: positive association
from clinical and in vitro analysis. Oral Oncol 2010; 46:
2048.
29. Chang KW, Liu CJ, Chu TH, et al. Association between
high miR-211 microRNA expression and the poor prognosis
of oral carcinoma. J Dent Res 2008; 87: 10638.
30. Liu CJ, Kao SY, Tu HF, Tsai MM, Chang KW, Lin SC.
Increase of microRNA miR-31 level in plasma could be a
potential marker of oral cancer. Oral Dis 2010; 16: 3604.

J Oral Pathol Med

31. Liu CJ, Lin SC, Yang CC, Cheng HW, Chang KW.
Exploiting salivary miR-31 as a clinical biomarker of oral
squamous cell carcinoma. Head Neck 2012; 34: 21924.
32. Hirono I. Natural carcinogenic products of plant origin. Crit
Rev Toxicol 1981; 8: 23577.
33. Chu NS. Effects of Betel chewing on the central and
autonomic nervous systems. J Biomed Sci 2001; 8: 22936.
34. Angadi PV, Rao SS. Areca nut in pathogenesis of oral
submucous brosis: revisited. Oral Maxillofac Surg 2011;
15: 19.
35. Chatrchaiwiwatana S. Dental caries and periodontitis associated with betel quid chewing: analysis of two data sets.
J Med Assoc Thai 2006; 89: 100411.
36. Tilakaratne WM, Klinikowski MF, Saku T, Peters TJ,
Warnakulasuriya S. Oral submucous brosis: review on
aetiology and pathogenesis. Oral Oncol 2006; 42: 5618.
37. Trivedy CR, Craig G, Warnakulasuriya S. The oral health
consequences of chewing areca nut. Addict Biol 2002; 7:
11525.
38. Mori H, Matsubara N, Ushimaru Y, Hirono I. Carcinogenicity examination of betel nuts and piper betel leaves.
Experientia 1979; 35: 3845.
39. Nair J, Nair UJ, Ohshima H, Bhide SV, Bartsch H.
Endogenous nitrosation in the oral cavity of chewers while
chewing betel quid with or without tobacco. IARC Sci Publ
1987; 84: 4659.
40. Wenke G, Brunnemann KD, Hoffmann D, Bhide SV. A
study of betel quid carcinogenesis. IV. Analysis of the saliva
of betel chewers: a preliminary report. J Cancer Res Clin
Oncol 1984; 108: 1103.
41. Atkinson L, Purohit R, Reay-Young P, Scott GC. Cancer
reporting in Papua New Guinea: 195870 and 197178. Natl
Cancer Inst Monogr 1982; 62: 6571.
42. Sharma DC. Betel quid and areca nut are carcinogenic
without tobacco. Lancet Oncol 2003; 4: 587.
43. Ashby J, Styles JA, Boyland E. Betel nuts, arecaidine, and
oral cancer. Lancet 1979; 1: 112.
44. Lin LM, Chen YK, Lai DR, Huang YL, Chen HR. Cancerpromoting effect of Taiwan betel quid in hamster buccal
pouch carcinogenesis. Oral Dis 1997; 3: 2325.
45. Liu TY, Chen CL, Chi CW. Oxidative damage to DNA
induced by areca nut extract. Mutat Res 1996; 367: 2531.
46. Nair UJ, Floyd RA, Nair J, Bussachini V, Friesen M, Bartsch
H. Formation of reactive oxygen species and of 8-hydroxydeoxyguanosine in DNA in vitro with betel quid ingredients. Chem Biol Interact 1987; 63: 15769.
47. Bhide SV, Shivapurkar NM, Gothoskar SV, Ranadive KJ.
Carcinogenicity of betel quid ingredients: feeding mice with
aqueous extract and the polyphenol fraction of betel nut. Br J
Cancer 1979; 40: 9226.
48. Lee CH, Lin RH, Liu SH, Lin-Shiau SY. Mutual interactions
among ingredients of betel quid in inducing genotoxicity on
Chinese hamster ovary cells. Mutat Res 1996; 367: 99104.
49. Panigrahi GB, Rao AR. Chromosome-breaking ability of
arecoline, a major betel-nut alkaloid, in mouse bone-marrow
cells in vivo. Mutat Res 1982; 103: 197204.
50. Wang YC, Tsai YS, Huang JL, et al. Arecoline arrests cells
at prometaphase by deregulating mitotic spindle assembly
and spindle assembly checkpoint: implication for carcinogenesis. Oral Oncol 2010; 46: 25562.
51. Tsai YS, Lee KW, Huang JL, et al. Arecoline, a major
alkaloid of areca nut, inhibits p53, represses DNA repair, and
triggers DNA damage response in human epithelial cells.
Toxicology 2008; 249: 2307.
52. Chang YC, Tai KW, Cheng MH, Chou LS, Chou MY.
Cytotoxic and non-genotoxic effects of arecoline on human

Overview of diet-mediated oral cancer


Li et al.

53.

54.

55.

56.
57.

58.
59.
60.
61.
62.

63.
64.

65.

66.
67.

68.

69.

70.

buccal broblasts in vitro. J Oral Pathol Med 1998; 27:


6871.
Chang YC, Hu CC, Tseng TH, Tai KW, Lii CK, Chou MY.
Synergistic effects of nicotine on arecoline-induced cytotoxicity in human buccal mucosal broblasts. J Oral Pathol
Med 2001; 30: 45864.
Chiang SL, Jiang SS, Wang YJ, et al. Characterization of
arecoline-induced effects on cytotoxicity in normal human
gingival broblasts by global gene expression proling.
Toxicol Sci 2007; 100: 6674.
Lin LM, Chen YK, Lai DL, Huang YL. Minimal arecaidine
concentrations showing a promotion effect during DMBAinduced hamster cheek pouch carcinogenesis. J Oral Pathol
Med 1996; 25: 658.
Dasgupta R, Saha I, Pal S, et al. Immunosuppression,
hepatotoxicity and depression of antioxidant status by
arecoline in albino mice. Toxicology 2006; 227: 94104.
Saha I, Chatterjee A, Mondal A, Maiti BR, Chatterji U.
Arecoline augments cellular proliferation in the prostate
gland of male Wistar rats. Toxicol Appl Pharmacol 2011;
255: 1608.
Jeng JH, Kuo ML, Hahn LJ, Kuo MY. Genotoxic and nongenotoxic effects of betel quid ingredients on oral mucosal
broblasts in vitro. J Dent Res 1994; 73: 10439.
Panigrahi GB, Rao AR. Study of the genotoxicity of the total
aqueous extract of betel nut and its tannin. Carcinogenesis
1986; 7: 379.
Liu CJ, Chen CL, Chang KW, Chu CH, Liu TY. Safrole in
betel quid may be a risk factor for hepatocellular carcinoma:
case report. CMAJ 2000; 162: 35960.
Chung YT, Chen CL, Wu CC, Chan SA, Chi CW, Liu TY.
Safrole-DNA adduct in hepatocellular carcinoma associated
with betel quid chewing. Toxicol Lett 2008; 183: 217.
Lee JM, Liu TY, Wu DC, et al. Safrole-DNA adducts in
tissues from esophageal cancer patients: clues to arecarelated esophageal carcinogenesis. Mutat Res 2005; 565:
1218.
Chen CL, Chi CW, Chang KW, Liu TY. Safrole-like DNA
adducts in oral tissue from oral cancer patients with a betel
quid chewing history. Carcinogenesis 1999; 20: 23314.
Ni WF, Tsai CH, Yang SF, Chang YC. Elevated expression
of NF-kappaB in oral submucous brosisevidence for NFkappaB induction by safrole in human buccal mucosal
broblasts. Oral Oncol 2007; 43: 55762.
Sundqvist K, Liu Y, Nair J, Bartsch H, Arvidson K,
Grafstrom RC. Cytotoxic and genotoxic effects of areca nutrelated compounds in cultured human buccal epithelial cells.
Cancer Res 1989; 49: 52948.
Lai KC, Lee TC. Genetic damage in cultured human
keratinocytes stressed by long-term exposure to areca nut
extracts. Mutat Res 2006; 599: 6675.
Lin SC, Liu CJ, Chiu CP, Chang SM, Lu SY, Chen YJ.
Establishment of OC3 oral carcinoma cell line and identication of NF-kappa B activation responses to areca nut
extract. J Oral Pathol Med 2004; 33: 7986.
Lu SY, Chang KW, Liu CJ, et al. Ripe areca nut extract
induces G1 phase arrests and senescence-associated
phenotypes in normal human oral keratinocyte. Carcinogenesis 2006; 27: 127384.
Ji WT, Yang SR, Chen JY, et al. Arecoline downregulates
levels of p21 and p27 through the reactive oxygen species/
mTOR complex 1 pathway and may contribute to oral
squamous cell carcinoma. Cancer Sci 2012; 103: 12219.
Costea DE, Hills A, Osman AH, et al. Identication of two
distinct carcinoma-associated broblast subtypes with differential tumor-promoting abilities in oral squamous cell
carcinoma. Cancer Res 2013; 73: 3888901.

71. Jung DW, Che ZM, Kim J, Kim K, Kim KY, Williams D.
Tumor-stromal crosstalk in invasion of oral squamous cell
carcinoma: a pivotal role of CCL7. Int J Cancer 2010; 127:
33244.
72. Yapijakis C, Bramos A, Nixon AM, Ragos V, Vairaktaris E.
The interplay between hemostasis and malignancy: the oral
cancer paradigm. Anticancer Res 2012; 32: 1791800.
73. Lu HH, Liu CJ, Liu TY, Kao SY, Lin SC, Chang KW.
Areca-treated broblasts enhance tumorigenesis of oral
epithelial cells. J Dent Res 2008; 87: 106974.
74. Tseng YH, Chang CS, Liu TY, Kao SY, Chang KW, Lin
SC. Areca nut extract treatment down-regulates involucrin in
normal human oral keratinocyte through P13K/AKT activation. Oral Oncol 2007; 43: 6709.
75. Liu SY, Liu YC, Huang WT, Huang GC, Chen TC, Lin MH.
Up-regulation of matrix metalloproteinase-8 by betel quid
extract and arecoline and its role in 2D motility. Oral Oncol
2007; 43: 102633.
76. Giri S, Poindexter KM, Sundar SN, Firestone GL. Arecoline
induced disruption of expression and localization of the tight
junctional protein ZO-1 is dependent on the HER 2
expression in human endometrial Ishikawa cells. BMC Cell
Biol 2010; 11: 53.
77. Deng YT, Chang JZ, Yeh CC, Cheng SJ, Kuo MY.
Arecoline stimulated Cyr61 production in human gingival
epithelial cells: inhibition by lovastatin. Oral Oncol 2011;
47: 25661.
78. Moutasim KA, Jenei V, Sapienza K, et al. Betel-derived
alkaloid up-regulates keratinocyte alphavbeta6 integrin
expression and promotes oral submucous brosis. J Pathol
2011; 223: 36677.
79. Shieh DH, Chiang LC, Shieh TY. Augmented mRNA
expression of tissue inhibitor of metalloproteinase-1 in
buccal mucosal broblasts by arecoline and safrole as a
possible pathogenesis for oral submucous brosis. Oral
Oncol 2003; 39: 72835.
80. Yang SC, Lin SC, Chiang WF, Yen CY, Lin CH, Liu SY.
Areca nut extract treatment elicits the broblastoid morphological changes, actin re-organization and signaling activation
in oral keratinocytes. J Oral Pathol Med 2003; 32: 6005.
81. Chiu CC, Chen BH, Hour TC, et al. Betel quid extract
promotes oral cancer cell migration by activating a muscarinic M4 receptor-mediated signaling cascade involving
SFKs and ERK1/2. Biochem Biophys Res Commun 2010;
399: 605.
82. Tseng YH, Chang KW, Yang CC, et al. Association
between areca-stimulated vimentin expression and the
progression of head and neck cancers. Head Neck 2012;
34: 24553.
83. Trivedy CR, Warnakulasuriya KA, Peters TJ, Senkus R,
Hazarey VK, Johnson NW. Raised tissue copper levels in
oral submucous brosis. J Oral Pathol Med 2000; 29:
2418.
84. Lu HH, Chen LK, Cheng CY, Hung SL, Lin SC, Chang
KW. Areca nut extract-treated gingival broblasts modulate
the invasiveness of polymorphonuclear leukocytes via the
production of MMP-2. J Oral Pathol Med 2009; 38: 7986.
85. Tran TN, Liu Y, Takagi M, Yamaguchi A, Fujii H.
Frequent promoter hypermethylation of RASSF1A and
p16INK4a and infrequent allelic loss other than 9p21 in
betel-associated oral carcinoma in a Vietnamese nonsmoking/non-drinking female population. J Oral Pathol
Med 2005; 34: 1506.
86. Lin PC, Chang WH, Chen YH, Lee CC, Lin YH, Chang JG.
Cytotoxic effects produced by arecoline correlated to
epigenetic regulation in human K-562 cells. J Toxicol
Environ Health A 2011; 74: 73745.

175

J Oral Pathol Med

Overview of diet-mediated oral cancer


Li et al.

176

87. Gilad S, Meiri E, Yogev Y, et al. Serum microRNAs are


promising novel biomarkers. PLoS ONE 2008; 3: e3148.
88. Mendell JT. MicroRNAs: critical regulators of development,
cellular physiology and malignancy. Cell Cycle 2005; 4:
117984.
89. Kong YW, Cannell IG, De Moor CH, et al. The mechanism
of micro-RNA-mediated translation repression is determined
by the promoter of the target gene. Proc Natl Acad Sci USA
2008; 105: 886671.
90. Baek D, Villen J, Shin C, Camargo FD, Gygi SP, Bartel DP.
The impact of microRNAs on protein output. Nature 2008;
455: 6471.
91. Bhattacharyya SN, Habermacher R, Martine U, Closs EI,
Filipowicz W. Relief of microRNA-mediated translational
repression in human cells subjected to stress. Cell 2006; 125:
111124.
92. Guo H, Ingolia NT, Weissman JS, Bartel DP. Mammalian
microRNAs predominantly act to decrease target mRNA
levels. Nature 2010; 466: 83540.
93. Miranda KC, Huynh T, Tay Y, et al. A pattern-based
method for the identication of MicroRNA binding sites and
their corresponding heteroduplexes. Cell 2006; 126:
120317.
94. Gomes CC, De Sousa SF, Gomez RS. MicroRNAs: small
molecules with a potentially role in oral squamous cell
carcinoma. Current Pharm Des 2013; 19: 128591.
95. Kantharidis P, Wang B, Carew RM, Lan HY. Diabetes
complications: the microRNA perspective. Diabetes 2011;
60: 18327.
96. Xu J, Zhao J, Evan G, Xiao C, Cheng Y, Xiao J. Circulating
microRNAs: novel biomarkers for cardiovascular diseases. J
Mol Med 2012; 90: 86575.
97. Wu BH, Xiong XP, Jia J, Zhang WF. MicroRNAs: new
actors in the oral cancer scene. Oral Oncol 2011; 47: 3149.
98. Szafranska AE, Davison TS, John J, et al. MicroRNA
expression alterations are linked to tumorigenesis and nonneoplastic processes in pancreatic ductal adenocarcinoma.
Oncogene 2007; 26: 444252.
99. Gomes CC, Gomez RS. MicroRNA and oral cancer: future
perspectives. Oral Oncol 2008; 44: 9104.
100. Perez-Sayans M, Pilar GD, Barros-Angueira F, et al. Current
trends in miRNAs and their relationship with oral squamous
cell carcinoma. J Oral Pathol Med 2012; 41: 43343.
101. Gorenchtein M, Poh CF, Saini R, Garnis C. MicroRNAs in
an oral cancer context from basic biology to clinical utility.
J Dent Res 2012; 91: 4406.
102. Lu HH, Kao SY, Liu TY, et al. Areca nut extract induced
oxidative stress and upregulated hypoxia inducing factor
leading to autophagy in oral cancer cells. Autophagy 2010;
6: 72537.
103. Liu CJ, Tsai MM, Hung PS, et al. miR-31 ablates expression
of the HIF regulatory factor FIH to activate the HIF pathway
in head and neck carcinoma. Cancer Res 2010; 70: 163544.
104. Gee HE, Camps C, Buffa FM, et al. hsa-mir-210 is a marker
of tumor hypoxia and a prognostic factor in head and neck
cancer. Cancer 2010; 116: 214858.
105. Tsai YS, Lin CS, Chiang SL, Lee CH, Lee KW, Ko YC.
Areca nut induces miR-23a and inhibits repair of DNA
double-strand breaks by targeting FANCG. Toxicol Sci
2011; 123: 48090.
106. Hung PS, Chang KW, Kao SY, Chu TH, Liu CJ, Lin SC.
Association between the rs2910164 polymorphism in premir-146a and oral carcinoma progression. Oral Oncol 2012;
48: 4048.
107. Kao SY, Chen YP, Tu HF, et al. Nuclear STK15 expression
is associated with aggressive behaviour of oral carcinoma
cells in vivo and in vitro. J Pathol 2010; 222: 99109.

J Oral Pathol Med

108. Ho TJ, Chiang CP, Hong CY, Kok SH, Kuo YS, Yen-Ping
Kuo M. Induction of the c-jun protooncogene expression by
areca nut extract and arecoline on oral mucosal broblasts.
Oral Oncol 2000; 36: 4326.
109. Hsieh LL, Wang PF, Chen IH, et al. Characteristics of
mutations in the p53 gene in oral squamous cell carcinoma
associated with betel quid chewing and cigarette smoking in
Taiwanese. Carcinogenesis 2001; 22: 1497503.
110. Jeng JH, Ho YS, Chan CP, et al. Areca nut extract upregulates prostaglandin production, cyclooxygenase-2
mRNA and protein expression of human oral keratinocytes.
Carcinogenesis 2000; 21: 136570.
111. Jeng JH, Wang YJ, Chiang BL, et al. Roles of keratinocyte
inammation in oral cancer: regulating the prostaglandin E2,
interleukin-6 and TNF-alpha production of oral epithelial
cells by areca nut extract and arecoline. Carcinogenesis
2003; 24: 130115.
112. Lin SC, Lu SY, Lee SY, Lin CY, Chen CH, Chang KW.
Areca (betel) nut extract activates mitogen-activated protein
kinases and NF-kappaB in oral keratinocytes. Int J Cancer
2005; 116: 52635.
113. Tseng YH, Chang KW, Liu CJ, Lin CY, Yang SC, Lin SC.
Areca nut extract represses migration and differentiation
while activating matrix metalloproteinase-9 of normal
gingival epithelial cells. J Periodontal Res 2008; 43: 4909.
114. Goldstein BY, Chang SC, Hashibe M, La Vecchia C, Zhang
ZF. Alcohol consumption and cancers of the oral cavity and
pharynx from 1988 to 2009: an update. Eur J Cancer Prev
2010; 19: 43165.
115. Zygogianni AG, Kyrgias G, Karakitsos P, et al. Oral
squamous cell cancer: early detection and the role of alcohol
and smoking. Head Neck Oncol 2011; 3: 2.
116. Humans IWGOTEOCRT. Tobacco smoke and involuntary
smoking. IARC Monogr Eval Carcinog Risks Hum 2004; 83:
11438.
117. Hecht SS. Tobacco carcinogens, their biomarkers and
tobacco-induced cancer. Nat Rev Cancer 2003; 3: 73344.
118. Boffetta P, Hashibe M. Alcohol and cancer. Lancet Oncol
2006; 7: 14956.
119. Hashibe M, Brennan P, Chuang SC, et al. Interaction
between tobacco and alcohol use and the risk of head and
neck cancer: pooled analysis in the International Head and
Neck Cancer Epidemiology Consortium. Cancer Epidemiol
Biomarkers Prev 2009; 18: 54150.
120. Brugere J, Guenel P, Leclerc A, Rodriguez J. Differential
effects of tobacco and alcohol in cancer of the larynx,
pharynx, and mouth. Cancer 1986; 57: 3915.
121. Znaor A, Brennan P, Gajalakshmi V, et al. Independent
and combined effects of tobacco smoking, chewing and
alcohol drinking on the risk of oral, pharyngeal and
esophageal cancers in Indian men. Int J Cancer 2003; 105:
6816.
122. Ko YC, Huang YL, Lee CH, Chen MJ, Lin LM, Tsai CC.
Betel quid chewing, cigarette smoking and alcohol consumption related to oral cancer in Taiwan. J Oral Pathol
Med 1995; 24: 4503.
123. Conney AH. Enzyme induction and dietary chemicals as
approaches to cancer chemoprevention: the Seventh DeWitt
S. Goodman Lecture. Cancer Res 2003; 63: 700531.
124. Khaf A, Schantz SP, Chou TC, Edelstein D, Sacks PG.
Quantitation of chemopreventive synergism between ()epigallocatechin-3-gallate and curcumin in normal, premalignant and malignant human oral epithelial cells. Carcinogenesis 1998; 19: 41924.
125. Lambert JD, Hong J, Yang GY, Liao J, Yang CS. Inhibition
of carcinogenesis by polyphenols: evidence from laboratory
investigations. Am J Clin Nutr 2005; 81: 284S91S.

Overview of diet-mediated oral cancer


Li et al.

126. Narotzki B, Reznick AZ, Navot-Mintzer D, Dagan B, Levy


Y. Green tea and vitamin E enhance exercise-induced
benets in body composition, glucose homeostasis, and
antioxidant status in elderly men and women. J Am Coll
Nutr 2013; 32: 3140.
127. Balakrishnan S, Manoharan S, Alias LM, Nirmal MR. Effect
of curcumin and ferulic acid on modulation of expression
pattern of p53 and bcl-2 proteins in 7,12-dimethylbenz[a]
anthracene-induced hamster buccal pouch carcinogenesis.
Indian J Biochem Biophys 2010; 47: 712.
128. Iriti M, Varoni EM. Chemopreventive potential of avonoids in oral squamous cell carcinoma in human studies.
Nutrients 2013; 5: 256476.
129. Shin HK, Kim J, Lee EJ, Kim SH. Inhibitory effect of
curcumin on motility of human oral squamous carcinoma
YD-10B cells via suppression of ERK and NF-kappaB
activations. Phytother Res 2010; 24: 57782.
130. Cheng SJ, Ko HH, Cheng SL, et al. Arecoline-stimulated
placenta growth factor production in gingival epithelial cells:
modulation by curcumin. Oral Dis 2013; 19: 51318.
131. Lee SS, Tsai CH, Ho YC, Chang YC. The upregulation of
heat shock protein 70 expression in areca quid chewingassociated oral squamous cell carcinomas. Oral Oncol 2008;
44: 88490.
132. Lee SS, Tsai CH, Yang SF, Ho YC, Chang YC. Hypoxia
inducible factor-1 alpha expression in areca quid chewingassociated oral squamous cell carcinomas. Oral Dis 2010;
16: 696701.
133. Yang CS, Wang ZY. Tea and cancer. J Natl Cancer Inst
1993; 85: 103849.
134. Lee UL, Choi SW. The chemopreventive properties and
therapeutic modulation of green tea polyphenols in oral
squamous cell carcinoma. ISRN Oncol 2011; 2011: 403707.
135. Lin SC, Li WC, Shih JW, Hong KF, Pan YR, Lin JJ. The tea
polyphenols EGCG and EGC repress mRNA expression of
human telomerase reverse transcriptase (hTERT) in carcinoma cells. Cancer Lett 2006; 236: 808.
136. Narotzki B, Reznick AZ, Aizenbud D, Levy Y. Green tea: a
promising natural product in oral health. Arch Oral Biol
2012; 57: 42935.
137. Li N, Chen X, Liao J, et al. Inhibition of 7,12-dimethylbenz[a]
anthracene (DMBA)-induced oral carcinogenesis in hamsters
by tea and curcumin. Carcinogenesis 2002; 23: 130713.
138. Srinivasan P, Suchalatha S, Babu PV, et al. Chemopreventive and therapeutic modulation of green tea polyphenols on drug metabolizing enzymes in 4-Nitroquinoline
1-oxide induced oral cancer. Chem Biol Interact 2008; 172:
22434.
139. Ide R, Fujino Y, Hoshiyama Y, et al. A prospective study of
green tea consumption and oral cancer incidence in Japan.
Ann Epidemiol 2007; 17: 8216.
140. Tsao AS, Liu D, Martin J, et al. Phase II randomized,
placebo-controlled trial of green tea extract in patients with
high-risk oral premalignant lesions. Cancer Prev Res 2009;
2: 93141.

141. Aggarwal BB, Sung B. Pharmacological basis for the role of


curcumin in chronic diseases: an age-old spice with modern
targets. Trends Pharmacol Sci 2009; 30: 8594.
142. Huang MT, Smart RC, Wong CQ, Conney AH. Inhibitory
effect of curcumin, chlorogenic acid, caffeic acid, and ferulic
acid on tumor promotion in mouse skin by 12-O-tetradecanoylphorbol-13-acetate. Cancer Res 1988; 48: 59416.
143. Rao CV, Simi B, Reddy BS. Inhibition by dietary curcumin
of azoxymethane-induced ornithine decarboxylase, tyrosine
protein kinase, arachidonic acid metabolism and aberrant
crypt foci formation in the rat colon. Carcinogenesis 1993;
14: 221925.
144. Anand P, Thomas SG, Kunnumakkara AB, et al. Biological
activities of curcumin and its analogues (Congeners) made
by man and Mother Nature. Biochem Pharmacol 2008; 76:
1590611.
145. Aggarwal BB, Harikumar KB. Potential therapeutic effects
of curcumin, the anti-inammatory agent, against neurodegenerative, cardiovascular, pulmonary, metabolic, autoimmune and neoplastic diseases. Int J Biochem Cell Biol 2009;
41: 4059.
146. Aggarwal BB, Kumar A, Bharti AC. Anticancer potential of
curcumin: preclinical and clinical studies. Anticancer Res
2003; 23: 36398.
147. Clark CA, Mceachern MD, Shah SH, et al. Curcumin
inhibits carcinogen and nicotine-induced Mammalian target
of rapamycin pathway activation in head and neck squamous
cell carcinoma. Cancer Prev Res (Phila) 2010; 3: 158695.
148. Balogun E, Hoque M, Gong P, et al. Curcumin activates the
haem oxygenase-1 gene via regulation of Nrf2 and the
antioxidant-responsive element. Biochem J 2003; 371:
88795.
149. Dudas J, Fullar A, Romani A, et al. Curcumin targets
broblast-tumor cell interactions in oral squamous cell
carcinoma. Exp Cell Res 2013; 319: 8009.
150. Kim JY, Cho TJ, Woo BH, et al. Curcumin-induced
autophagy contributes to the decreased survival of oral
cancer cells. Arch Oral Biol 2012; 57: 101825.
151. Thangapazham RL, Sharma A, Maheshwari RK. Multiple
molecular targets in cancer chemoprevention by curcumin.
AAPS J 2006; 8: E4439.
152. Yu CC, Tsai LL, Wang ML, et al. miR145 targets the
SOX9/ADAM17 axis to inhibit tumor-initiating cells and
IL-6-mediated paracrine effects in head and neck cancer.
Cancer Res 2013; 73: 342540.

177

Acknowledgements
This work is supported by Grants NSC102-2314-B-010-010 and NSC102-2628B-010-011-MY3 from National Science Council, Taiwan, as well as Grants
99TPECH06 and 100TPECH06 from Taipei City Hospital/Department of
Health, Taipei City Government. We also would like to thank Ms Courtney
Anne Curtis for critical review and English edition for the manuscript.

J Oral Pathol Med

You might also like