DNA Methylation As An Epigenetic Regulator of Gallbladder Cancer - An Overview

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

International Journal of Surgery 53 (2018) 178–183

Contents lists available at ScienceDirect

International Journal of Surgery


journal homepage: www.elsevier.com/locate/ijsu

Review

DNA methylation as an epigenetic regulator of gallbladder cancer: An T


overview
Jibran Sualeh Muhammada,b,∗, Muhammad Rizwan Khanc, Kulsoom Ghiasb
a
Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
b
Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
c
Department of Surgery, The Aga Khan University, Karachi, Pakistan

A R T I C LE I N FO A B S T R A C T

Keywords: Gallbladder cancer (GBC) is a lethal health issue affecting mostly the women in their middle-age. High incidence
Gallbladder of GBC has been reported across the world specifically in Asian countries, India and Pakistan. The exact etiology
Gallbladder cancer remains unknown, although several risk factors and genetic aberrations involving mutations or epigenetic
Epigenetic regulation changes may be involved in gallbladder carcinogenesis. This article presents a review of the published literature
DNA methylation
mainly from the year 2003 onwards. The topic of main concerns was epigenetic regulation of GBC. All relevant
studies identified were included and are described according to the aforementioned subheadings. In this review,
we have discussed the role of DNA methylation in GBC, clinical implication and future prospects of biomarker
development for early diagnosis and therapeutic interventions.

1. Introduction mechanisms, DNA methylation has been most extensively studied with
regards to regulation of carcinogenesis [8]. DNA methylation results in
Gallbladder cancer (GBC) is a rare, but often fatal malignancy with reversible changes and therefore are an attractive therapeutic target for
ethnic, geographic, and socioeconomic variation in its incidence. cancer treatment. From the year 2003 onwards, several studies on the
Recent statistics reveals greater than 10,000 new cases and over 3000 role of DNA methylation in GBC were reported for global populations,
deaths per year due to GBC in the United States alone [1]. High in- including from United States, Chile, and several Asian countries. Most
cidence of GBC has also been reported in South American and South of these studies were performed on Chilean population, reporting hy-
Asian countries, including India and Pakistan [2–4]. More than 70% of permethylation in genes related to cell adhesion, cell cycle, and p53
GBC patients are female, making it one of the most common malig- pathways. On the other hand, studies from Indian population showed a
nancies of the gastrointestinal tract in women from these regions [2–6]. significant role of increased promoter methylation of tumor suppressor
Some of the cases of GBC are found incidentally in patients undergoing genes with respect to advance stages of GBC [9,10]. Hence, high in-
cholecystectomy for the treatment of cholecystitis or cholelithiasis, but cidence and poor prognosis of GBC reported in these Asian and Amer-
most of the patients usually presents late with subtle nonspecific ican countries was directly related to gene regulation via promoter
symptoms. By then, the cancer has usually reached at an advanced hypermethylation. In this systematic review, we present the highlights
stage and possesses a high risk of metastases. Complete removal of the of relevant studies reporting promoter methylation of specific genes
gallbladder with adjacent liver parenchyma and the draining lymph involved in epigenetic regulation of GBC. Based on the available lit-
nodes by a specialized surgeon is the only curative treatment option, erature, we also provide an insight into clinical implications, ther-
which may not be possible in advanced cases. Research on GBC has apeutic approaches and future directions in the field.
therefore been focused on identifying biomarkers for early diagnosis
and molecular targets for therapeutic interventions. 2. Methods
One particular area of focus is epigenetics, which may play a role in
disease onset and progression. Epigenetic mechanisms, such as DNA We used the MeSH terms “gallbladder” and “gallbladder cancer”
methylation, histone modification, and regulation of gene expression by with “epigenetics,” or “DNA methylation,” to search the scientific da-
microRNA (miRNA), are defined as involving changes to the phenotype tabases, such as SCOPUS, Ovid, PubMed and Web of science. To identify
without any change in DNA sequence itself [7]. Of these key epigenetic additional studies, we hand-searched the reference lists of the studies


Corresponding author. Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
E-mail address: dr.jibran@live.com (J.S. Muhammad).

https://doi.org/10.1016/j.ijsu.2018.03.053
Received 14 August 2017; Received in revised form 20 March 2018; Accepted 21 March 2018
Available online 27 March 2018
1743-9191/ © 2018 IJS Publishing Group Ltd. Published by Elsevier Ltd. All rights reserved.
J.S. Muhammad et al. International Journal of Surgery 53 (2018) 178–183

Fig. 1. Flowchart outlining the search strategy employed to identify the relevant studies.

identified using MeSH terms. The flowchart in Fig. 1 depicts the search gastric epithelial cells [15]. With regards to GBC, the role of DNA
strategy employed to identify relevant studies. methylation has been extensively studied as compared to histone
modification. Only a few studies have reported differential expression
of miRNAs in GBC [16–18]. However, solid evidence is still lacking with
3. Epigenetic regulation of carcinogenesis respect to any link between methylation of miRNA or histone mod-
ifications in the development of gallbladder carcinogenesis. Therefore,
Epigenetic alterations associated with cancer development includes the main focus of this review is the role of DNA methylation in gall-
silencing of tumor suppressor genes via methylation, inactivation of bladder carcinogenesis.
DNA repair genes by histone acetylation and miRNA regulation of cell
proliferation, differentiation, and apoptosis [11]. Aberrant epigenetic
changes in the DNA causing alteration and deregulation of the normal 4. DNA methylation as an epigenetic regulator of gallbladder
cell cycle processes leads to abnormal cell survival and initiation and cancer
progression of carcinogenesis. DNA methylation involves the conver-
sion of the nucleotide cytosine, specifically found before guanine in Chronic inflammation of the gallbladder as a consequence of gall
CpG islands, into methylcytosine [12]. The promoter region of nu- stones or bacterial infections is a well-documented risk factor for GBC.
merous tumor suppressor genes and DNA repair genes contains CpG Chronic cellular irritation might be responsible for activation of in-
islands and are, therefore, highly prone to methylation. Hypermethy- tracellular enzymes involved in hypermethylation of the promoter re-
lation of the promoter region of a gene directly blocks the binding of gion of tumor suppressor or cell cycle regulating genes. These epige-
transcription factors essential for gene expression, thus rendering loss of netic changes, in turn, contribute to early-stage tumorigenesis in GBC
expression or gene silencing [13]. Studies have reported higher me- and are also seen in cholelithiasis. The methylation profile of previously
thylation levels of various tumor suppressor genes, such as APC, reported genes and the functional significance of the epigenetic changes
BRCA1, and Long interspersed nuclear elements (LINE-1), which are in GBC are summarized in Table 1. A perspective figure explaining the
associated with the development of breast cancer [14]. Hypermethy- association of Gallbladder stones, and chronic inflammation leading to
lation of other genes besides tumor suppressor genes has also been DNA methylation of specific genes is shown (Fig. 2).
strongly linked with cancer initiation and progression. A recent study
showed that hypermethylation-induced silencing of an autophagy-re-
lated gene was directly associated with carcinogenic transformation in

179
J.S. Muhammad et al. International Journal of Surgery 53 (2018) 178–183

Table 1
Description of the genes frequently found methylated in gallbladder cancer.
Gene Gene function (http://www.genecards.org/) Reference

APC Encodes a tumor suppressor protein and involved in cell migration, cell adhesion, transcriptional activation, and apoptosis [9, 10, 19–22]
SHP1 Regulate a variety of cellular processes including cell growth, differentiation, mitotic cycle, and oncogenic transformation [10]
3-OST-2 Gene knockdown increases cell proliferation, invasion and migration [10]
CDH1 & CDH13 Loss of function of these genes is thought to contribute to cancer progression by increasing proliferation, invasion, and/or [10, 19–21, 27]
metastasis
PTEN Functions as a tumor suppressor by negatively regulating AKT/PKB signaling pathway and regulation of cell cycle and cell cycle [31, 32]
progression
CDKN2A/p16 Cell cycle regulation by slowing the progress of dividing cells from G1 to S phase [10, 19–22, 34, 35, 37, 38]
WIF-1 This gene functions as a tumor suppressor gene, and has been found to be epigenetically silenced in various cancers [40, 41]

4.1. Adenomatous polyposis coli (APC) (5q22.2) aberrant promoter methylation in gallbladder tissue samples from pa-
tients with chronic cholecystitis without cancer and tissue samples from
Tekchamen et al. reported hypermethylation and subsequent down- GBC patients. The MS-PCR and combined restriction analysis methods
regulation of the adenomatous polyposis coli (APC) gene promoter in were used to detect methylation. The study revealed significantly
96% of GBC patient samples studied in the Indian population. higher methylation frequencies in several genes in the cancer samples
Methylation-specific (MS)-PCR was used to analyze the methylation of as compared to cholecystitis samples. Of genes tested, SHP1 methyla-
APC gene. The expression of APC gene was studied by semi-quantitative tion was highest in GBC in 80% of tumor samples [10]. Interestingly,
PCR, real-time PCR and immunohistochemistry. Although methylation SHP1 was also methylated in 88% of chronic cholecystitis samples.
was also observed in 80% of the gallbladder tissue samples from pa- SHP1 is a tyrosine phosphatase known to regulate cell growth and
tients with gallstone disease, functional loss of APC was seen more in apoptosis [25]. While abnormal SHP1 expression has been reported in
GBC suggesting that methylation silencing of APC may play a role in the various cancers [26], its significance has not been explicitly studied in
early stages of gallbladder carcinogenesis [9]. Studies from Chile also GBC. Hypothetically, hypermethylation and silencing of SHP1 in GBC
reported methylation of the APC gene promoter region [19–22], with a could lead to increased cell proliferation. The same study also reported
progressive increase in the level of the methylation status of several higher methylation frequency of 3-O-sulfotransferase 2 (3-OST-2) gene
genes from chronic cholecystitis to advanced carcinoma. Functional in GBC samples as compared with cholecystitis samples [26]. Previous
loss of APC is known to activate Wnt signaling pathway and an increase studies in breast cancer have suggested a role for 3-OST-2 in the pro-
in the levels of the transcription factor β-catenin inside the nucleus, gression to malignant cells in early stages of the diseases [27], with
resulting in cellular proliferation [24]. The functional significance of gene knockdown strongly linked to increased cell proliferation, inva-
methylation of APC has been studied extensively in colorectal carci- sion, and migration. Similar to SHP1, a decrease in expression of 3-OST-
noma, and loss of APC secondary to chronic inflammation due to 2 is hypothesized to be involved in gallbladder carcinogenesis.
gallstones may be similarly responsible for GBC carcinogenesis.
4.3. CDH1 (16q22.1)
4.2. SHP1 (12p13.31)
This gene is responsible for encoding the Cadherin family of pro-
This protein encoded by PTPN6 gene is known to modulate sig- teins essential for regulating cell-cell adhesions by interacting with
naling by tyrosine phosphorylated cell surface receptors such as KIT themselves in a homophilic manner, and are frequently hypermethy-
and the EGFR. Takahashi et al. evaluated 24 tumor suppressor genes for lated in many types of cancers. CDH1, which encodes for a protein

Fig. 2. A perspective figure showing association of Gallbladder stones, and chronic inflammation leading to DNA methylation of specific genes.

180
J.S. Muhammad et al. International Journal of Surgery 53 (2018) 178–183

called E-cadherin, was found to be methylated to varying degrees in 4.7. Others


GBC in different populations, specifically in 41% of Japanese samples
[28] and in more than 65% of Chilean patients [19–21,27]. Another FHIT, DUTT1, BLU, and SEMA3B, genes present on the short arm of
gene related to cell adhesion, CDH13, was found to be hypermethylated chromosome 3 and involved in cell cycle regulation and DNA repair
in up to 80% of gallbladder cancer patients [11,20]. Epigenetic silen- regulation, have been reported to be associated with several epithelial
cing, detected using MS-PCR, or loss of function of both these genes cancers. Riquelme et al. [42] hypothesized that aberrant methylation of
contributes to cancer progression by increasing proliferation, migra- the CpG islands of the promoter region of these genes might also be
tion, invasion, and metastasis. linked with gallbladder carcinogenesis. All four genes were found to be
hypermethylated; 92% of surgically resected GBC cases exhibited me-
4.4. PTEN (10q23.31) thylation in the promoter region of SEMA3B, 66% of cases in FHIT, 26%
in BLU and 22% in DUTT1, suggesting that these tumor suppressor
Another tumor suppressor gene, PTEN, is often found to be silenced genes are a more fragile target for aberrant promoter methylation
by methylation in several cancers, including breast cancer [29]. PTEN contributing to the pathogenesis of GBC.
negatively regulates intracellular levels of phosphatidylinositol-3,4,5- Several other genes have also been reported to be methylated in
trisphosphate in cells and functions as a tumor suppressor by inhibiting GBC samples with intermediate to low frequencies. Gracia et al. [20]
the AKT/PKB signaling pathway [30]. Ali et al. recruited 141 con- analyzed both pre-neoplastic and neoplastic gallbladder tissue samples
secutive GBC patients in India, of which 48% showed hypermethylation in the Chilean population for methylation of DLC1, DAPK1, MGMT,
of the promoter region of PTEN, using MS-PCR. The methylation status TIMP3 and RARβ2 genes. Of these, only DLC1 gene methylation was
was not associated with the level of metaplasia and was similar in all more common in invasive cancers and was associated with poor sur-
stages of GBC. Immunohistochemical analysis revealed decreased PTEN vival [20]. In a study in a Korean population, frequent methylation was
expression in almost all methylated samples [31]. In another study from found in RASSF1 gene (22.7%) in GBC patients [43], prompting the
India, Tekcham et al. [32] showed PTEN promoter methylation in 30% authors to infer a possible role in gallbladder cancer development. In
of GBC patients and in 22% of patients with gallstones. RT-PCR analysis another Korean study [44], PGP9.5 promoter hypomethylation was
of PTEN in advanced GBC cases revealed significantly lower expression, found to be a reliable marker for GBC development. Another novel
especially in grade III cancer samples compared to those of other genes, tumor suppressor gene, cysteine dioxygenase type 1 (CDO1) gene was
suggesting that PTEN promoter methylation could be a useful bio- recently reported to be epigenetically regulated in GBC. These findings
marker for diagnosis or prognosis of GBC. suggested that CDO1 methylation was significantly associated with the
phenotypic acquisition of invasive and metastatic properties in primary
4.5. Cyclin dependent kinase inhibitor 2A (CDKN2A)/p16 (9q21.3) GBC. Notably, CDO1 hypermethylation showed prognostic relevance,
especially in stage II GBC, in which it is highly anticipated to work as a
CDKN2A, commonly known as p16, is a tumor suppressor gene predictive marker for candidates of adjuvant therapy [45].
frequently mutated or deleted in a wide variety of cancers. p16 plays a
key role in regulation of the cell cycle, specifically by interacting with 5. Clinical implications and future directions
CDK4 and CDK6, inhibiting their ability to interact with Cyclin D, hence
slowing the progression of rapidly dividing cells from G1 to S phase. Gallbladder carcinogenesis is usually considered a multistep process
Genetic or epigenetic reactivation of p16, therefore, is a potential ap- attributed to both genetic and environmental factors. The carcinogenic
proach for cancer therapy [33]. Of the 51 cancerous gallbladder spe- models in the development of gallbladder cancer have been well de-
cimens collected and analyzed in the Japanese population, Tadokoro scribed in the past and include the metaplasia-dysplasia-carcinoma se-
et al. reported methylation and loss of expression of p16 in 73% and quence or the adenoma-carcinoma sequence [46]. There is some clin-
62% of cases, respectively [34]. Decreased expression was attributed ical and experimental data to support both the models, but severe
either to loss of allele heterozygosity or promoter hypermethylation, as dysplasia has been found in most of the cases of gallbladder cancer
no p16 mutations were detected. While the sample size was limited, suggesting that the former sequence is a frequent pathway [47]. There
another study also showed p16 gene promoter methylation in 22% of is also some data from Pakistan indicating that approximately 40% of
GBC patients [35]. Several studies from Chile [11,19–22,36], Germany the patients with gallstones have intestinal metaplasia of the gall-
[37] and China [38] have investigated the role of p16 and reported bladder epithelium, which is considered a precursor lesion for gall-
varying frequency of gene hypermethylation, ranging from 20% to 60% bladder cancer [6]. Previous studies suggest a long latency period for
in GBC patients. All studies have used MS-PCR to analyze methylation gallbladder carcinogenesis and approximately 15 years may be required
levels. Overall, methylation-induced loss of p16 gene expression is for progression from dysplasia to advanced carcinoma [48,49]. There-
significantly associated with various premalignant and malignant le- fore, efforts are needed to identify the genetic and environmental fac-
sions in GBC and could be an early event in cancer progression [23]. tors responsible for the development of precursor lesions many years
before the appearance of cancer. Epigenetic changes, if detected early
4.6. WIF1 (12q14.3) in the course of the disease, may help in early diagnosis of the gall-
bladder cancer resulting in improved prognoses.
The Wnt inhibitory factor 1 gene is an inhibitor of the Wnt pathway. Early detection of epigenetic changes in gallbladder cancer may
Abnormal activation of the Wnt pathway is known to increase cell help improve the clinical outcomes of the patients with gallbladder
proliferation and differentiation, leading to tumor development [39]. A cancer in a number of ways. Overall, a number of clinical applications
study in a gallbladder cancer cell line showed a low expression of WIF- of epigenetic changes have been suggested in the literature. Fragments
1, which was associated with promoter methylation of the WIF1 gene of genomic DNA released from the necrotic tumor cells may enter the
[40]. Treatment of the GB-SD cells with a demethylating agent restored circulation or be released in the body fluids where they can be used as
expression of WIF1. Additionally, transfection with WIF1 expression biomarkers for the diagnosis, staging, or post-treatment monitoring of
plasmid inhibited cell proliferation, invasion, and metastasis accom- cancer [50]. The epigenetic changes may be used as a marker of ade-
panied by increased expression of matrix metalloproteinase. Another quate surgical resection according to gene methylation profile of the
study has shown that the level of WIF1 protein expression is low in WIF- tumor tissues compared to normal tissues including the tissues at the
1 hypermethylated specimens and multivariate analysis revealed that margin of resection [51]. Moreover, epigenetic changes may also be
WIF-1 methylation was an independent prognostic factor for 5-year used as a marker of response to local as well as systemic treatment [52].
overall survival [41]. The limiting factor is that such epigenetic markers are still in the

181
J.S. Muhammad et al. International Journal of Surgery 53 (2018) 178–183

experimental phase for gallbladder cancer, and there is no available References


data on their clinical use. One potential factor limiting the development
of such markers is the non-uniform geographic distribution of the dis- [1] R.L. Siegel, K.D. Miller, A. Jemal, Cancer statistics, 2015, CA A Cancer J. Clin. 65
ease itself. Variable data reviewed in this study from different popula- (1) (2015 Jan-Feb) 5–29, http://dx.doi.org/10.3322/caac.21254.
[2] L.M. Stinton, E.A. Shaffer, Epidemiology of gallbladder disease: cholelithiasis and
tions revealed no universal epigenetic biomarker. Additional work is cancer, Gut Liver 6 (2) (2012 Apr) 172–187.
required in high-risk regions to identify population-specific epigenetic [3] C. Are, H. Ahmad, A. Ravipati, D. Croo, D. Clarey, L. Smith, R.R. Price, J.M. Butte,
markers with enhanced clinical outcomes for patients. S. Gupta, A. Chaturvedi, S. Chowdhury, Global epidemiological trends and varia-
tions in the burden of gallbladder cancer, J. Surg. Oncol. 115 (5) (2017) 580–590.
Gallbladder cancer remains a fatal disease as it is usually diagnosed [4] Y. Bhurgri, A. Bhugri, S.H. Hassan, Cancer incidents in karachi Pakistan: first results
at very late stages and there are no specific epigenetic biomarkers in from karachi cancer registry, Int. J. Canc. 85 (2000) 325–329.
consideration. Probably combination of the several genes discussed in [5] A.R. Alvi, N.A. Siddiqui, H. Zafar, Risk factors of gallbladder cancer in Karachi-a
case-control study, World J. Surg. Oncol. 9 (2011 Dec 9) 164.
this review may prove to be an effective biomarker. However, a novel [6] M.R. Khan, S.A. Raza, Z. Ahmad, S. Naeem, S. Pervez, A.A. Siddiqui, M. Ahmed,
and population-specific biomarker facilitating early diagnosis, and later R. Azami, Gallbladder intestinal metaplasia in Pakistani patients with gallstones,
enabling prognostic monitoring are urgently required. Epigenetic Int. J. Surg. 9 (6) (2011) 482–485.
[7] C. Dupont, D.R. Armant, C.A. Brenner, Epigenetics: definition, mechanisms and
modifications of the DNA are hopeful for the development of bio-
clinical perspective, Semin. Reprod. Med. 27 (5) (2009 Sep) 351–357.
markers for specific cancers. [8] Q.W. Chen, X.Y. Zhu, Y.Y. Li, Z.Q. Meng, Epigenetic regulation and cancer (review),
Here we have summarized the role of several genes that are fre- Oncol. Rep. 31 (2) (2014 Feb) 523–532.
quently methylated in GBC and described the relationship of methyla- [9] D.S. Tekcham, S.S. Poojary, S. Bhunia, M.A. Barbhuiya, S. Gupta, B.R. Shrivastav,
P.K. Tiwari, Adenomatous polylosis coli (APC) gallbladder: epigenetic regulation of
tion of various gene promoters to disease development in specific po- APC in the molecular pathogenesis of gallbladder cancer, Indian J. Med. Res. 143
pulations. We regard the utility of immunohistochemistry in the (S1) (2016) 82–90.
expression of specific proteins in diagnosing and predicting prognosis of [10] T. Takahashi, N. Shivapurkar, E. Riquelme, H. Shigematsu, J. Reddy, M. Suzuki,
K. Miyajima, X. Zhou, B.N. Bekele, A.F. Gazdar, Wistuba II, Aberrant promoter
GBC, however, promoter methylation levels could be detected from hypermethylation of multiple genes in gallbladder carcinoma and chronic chole-
genomic DNA obtained from the blood plasma of GBC patients em- cystitis, Clin. Canc. Res. 10 (2004 Sep 15) 6126–6133 (18 Pt 1).
phasizing its use as less invasive diagnostic and prognostic applications. [11] Y. Tian, Q. Xie, J. He, X. Luo, T. Zhou, Y. Liu, Z. Huang, Y. Tian, D. Sun, K. Yao,
Radioactive (125)I seeds inhibit cell growth and epithelial-mesenchymal transition
Identification of population-specific epigenetic biomarkers in genomic in human glioblastoma multiforme via a ROS-mediated signaling pathway, BMC
DNA and prospective correlation with clinical outcomes has the po- Canc. 15 (2015 Feb 19) 1.
tential to further aid in earlier diagnosis and management of gall- [12] M. Esteller, Epigenetics in cancer, N. Engl. J. Med. 358 (11) (2008 Mar 13)
1148–1159.
bladder cancer. [13] P.A. Jones, D. Takai, The role of DNA methylation in mammalian epigenetics,
Science 293 (5532) (2001 Aug 10) 1068–1070.
[14] R. Radpour, Z. Barekati, C. Kohler, Q. Lv, N. Bürki, C. Diesch, J. Bitzer, H. Zheng,
Ethical approval S. Schmid, X.Y. Zhong, Hypermethylation of tumor suppressor genes involved in
critical regulatory pathways for developing a blood-based test in breast cancer,
Not applicable. PLoS One 6 (1) (2011 Jan 24) e16080.
[15] J.S. Muhammad, S. Nanjo, T. Ando, S. Yamashita, T. Maekita, T. Ushijima,
Y. Tabuchi, T. Sugiyama, Autophagy impairment by Helicobacter pylori-induced
methylation silencing of MAP1LC3Av1 promotes gastric carcinogenesis, Int. J.
Sources of funding
Canc. 140 (10) (2017) 2272–2283.
[16] G. Li, Y. Pu, MicroRNA signatures in total peripheral blood of gallbladder cancer
Not applicable. patients, Tumor Biol. 36 (2015) 6985–6990.
[17] B. Yang, B. Liu, P. Bi, T. Wu, Q. Wang, J. Zhang, An integrated analysis of differ-
ential miRNA and mRNA expressions in human gallstones, Mol. Biosyst. 11 (2015)
Author contribution 1004–1011.
[18] P. Letelier, P. Garcia, P. Leal, et al., miR-1 and miR-145 act as tumor suppressor
microRNAs in gallbladder cancer, Int. J. Clin. Exp. Pathol. 7 (2014) 1849–1867.
JSM, MRK and KG conceived the idea and planned the study. JSM [19] J.C. Roa, L. Anabalón, I. Roa, A. Melo, J.C. Araya, O. Tapia, X. de Aretxabala,
performed literature search, designed study methodology and drafted S. Muñoz, B. Schneider, Promoter methylation profile in gallbladder cancer, J.
Gastroenterol. 41 (3) (2006 Mar) 269–275.
the initial manuscript including the table and figure. MRK and KG [20] P. García, C. Manterola, J.C. Araya, M. Villaseca, P. Guzmán, A. Sanhueza,
critically analyzed and revised the manuscript. Final manuscript was M. Thomas, H. Alvarez, J.C. Roa, Promoter methylation profile in preneoplastic and
approved by all authors. neoplastic gallbladder lesions, Mol. Carcinog. 48 (1) (2009 Jan) 79–89.
[21] S.J.C. Roa, M.P. García, A.A. Melo, E.O. Tapia, H.M. Villaseca, O.J.C. Araya,
G.P. Guzmán, Gene methylation patterns in digestive tumors, Rev. Med. Chile 136
Conflicts of interest (4) (2008 Apr) 451–458.
[22] L.T. Kagohara, J.L. Schussel, T. Subbannayya, N. Sahasrabuddhe, C. Lebron,
M. Brait, L. Maldonado, B.L. Valle, F. Pirini, M. Jahuira, J. Lopez, P. Letelier,
None. P. Brebi-Mieville, C. Ili, A. Pandey, A. Chatterjee, D. Sidransky, R. Guerrero-Preston,
Global and gene-specific DNA methylation pattern discriminates cholecystitis from
gallbladder cancer patients in Chile, Future Oncol. 11 (2) (2015) 233–249.
Research registration Unique Identifying Number (UIN) [23] M.G. House, Wistuba II, P. Argani, M. Guo, R.D. Schulick, R.H. Hruban,
J.G. Herman, A. Maitra, Progression of gene hypermethylation in gallstone disease
leading to gallbladder cancer, Ann. Surg Oncol. 10 (8) (2003 Oct) 882–889.
Not applicable. [24] A.B. Sparks, P.J. Morin, B. Vogelstein, K.W. Kinzler, Mutational analysis of the APC/
beta-catenin/Tcf pathway in colorectal cancer, Canc. Res. 58 (6) (1998 Mar 15)
1130–1134.
Guarantor [25] M.D. Christensen, C. Geisler, Recruitment of SHP-1 protein tyrosine phosphatase
and signalling by a chimeric T-cell receptor-killer inhibitory receptor, Scand. J.
Immunol. 51 (6) (2000 Jun) 557–564.
Dr. Jibran Sualeh Muhammad MBBS; Ph.D. [26] C. Wu, M. Sun, L. Liu, G.W. Zhou, The function of the protein tyrosine phosphatase
Assistant Professor. SHP-1 in cancer, Gene 306 (2003 Mar 13) 1–12.
Department of Basic Medical Sciences, College of Medicine, [27] D. Dietrich, R. Lesche, R. Tetzner, M. Krispin, J. Dietrich, W. Haedicke, M. Schuster,
G. Kristiansen, Analysis of DNA methylation of multiple genes in microdissected
University of Sharjah, Sharjah, United Arab Emirates. cells from formalin-fixed and paraffin-embedded tissues, J. Histochem. Cytochem.
Email: dr.jibran@live.com. 57 (5) (2009 May) 477–489.
[28] Y. Koga, Y. Kitajima, A. Miyoshi, K. Sato, K. Kitahara, H. Soejima, K. Miyazaki,
Tumor progression through epigenetic gene silencing of O(6)-methylguanine-DNA
Disclosure methyltransferase in human biliary tract cancers, Ann. Surg Oncol. 12 (5) (2005)
354–363.
[29] S. Siddiqui, N. Akhter, S.V. Deo, N.K. Shukla, S.A. Husain, A study on promoter
None.

182
J.S. Muhammad et al. International Journal of Surgery 53 (2018) 178–183

methylation of PTEN in sporadic breast cancer patients from North India, Breast cancer tumorigenesis that predicts poor survival, Gene 622 (2017 Jul 30) 42–49.
Canc. 23 (6) (2016 Nov) 922–931. [42] E. Riquelme, M. Tang, S. Baez, A. Diaz, M. Pruyas, Wistuba II, A. Corvalan, Frequent
[30] G. Tamura, Alterations of tumor suppressor and tumor-related genes in the devel- epigenetic inactivation of chromosome 3p candidate tumor suppressor genes in
opment and progression of gastric cancer, World J. Gastroenterol. 12 (2) (2006 Jan gallbladder carcinoma, Canc. Lett. 250 (1) (2007 May 18) 100–106.
14) 192–198. [43] S.K. Kee, J.Y. Lee, M.J. Kim, S.M. Lee, Y.W. Jung, Y.J. Kim, J.Y. Park, H.I. Bae,
[31] A. Ali, P.K. Mishra, S. Sharma, A. Arora, S.S. Saluja, Effects of PTEN gene alteration H.S. Hong, Y.K. Yun, S.G. Kim, D.S. Kim, Hypermethylation of the Ras association
in patients with gallbladder cancer, Cancer Genet. 208 (12) (2015 Dec) 587–594. domain family 1A (RASSF1A) gene in gallbladder cancer, Mol. Cell. 24 (3) (2007
[32] D.S. Tekcham, S. Gupta, B.R. Shrivastav, P.K. Tiwari, Epigenetic downregulation of Dec 31) 364–371.
PTEN in gallbladder cancer, J. Gastrointest. Canc. 48 (1) (2017 Mar) 110–116. [44] Y.M. Lee, J.Y. Lee, M.J. Kim, H.I. Bae, J.Y. Park, S.G. Kim, D.S. Kim,
[33] R. Zhao, B.Y. Choi, M.H. Lee, A.M. Bode, Z. Dong, Implications of genetic and Hypomethylation of the protein gene product 9.5 promoter region in gallbladder
epigenetic alterations of CDKN2A (p16(INK4a)) in cancer, EBioMedicine 8 (2016 cancer and its relationship with clinicopathological features, Canc. Sci. 97 (11)
Jun) 30–39. (2006 Nov) 1205–1210.
[34] H. Tadokoro, T. Shigihara, T. Ikeda, M. Takase, M. Suyama, Two distinct pathways [45] K. Igarashi, K. Yamashita, H. Katoh, K. Kojima, Y. Ooizumi, N. Nishizawa,
of p16 gene inactivation in gallbladder cancer, World J. Gastroenterol. 13 (47) R. Nishiyama, H. Kawamata, H.1 Tajima, T. Kaizu, Y. Kumamoto, M. Watanabe,
(2007 Dec 21) 6396–6403. Prognostic significance of promoter DNA hypermethylation of the cysteine dioxy-
[35] T. Tozawa, G. Tamura, T. Honda, S. Nawata, W. Kimura, N. Makino, S. Kawata, genase 1 (CDO1) gene in primary gallbladder cancer and gallbladder disease, PLoS
T. Sugai, T. Suto, T. Motoyama, Promoter hypermethylation of DAP-kinase is as- One 12 (11) (2017 Nov 21) e0188178.
sociated with poor survival in primary biliary tract carcinoma patients, Canc. Sci. [46] P. Letelier, P. Brebi, O. Tapia, J.C. Roa, DNA promoter methylation as a diagnostic
95 (9) (2004 Sep) 736–740. and therapeutic biomarker in gallbladder cancer, Clin. Epigenet. 4 (1) (2012 Jul
[36] J.C. Roa, Q. Vo, J.C. Araya, M. Villaseca, P. Guzmán, G.S. Ibacache, X. de 13) 11.
Aretxabala, I. Roa, Inactivation of CDKN2A gene (p16) in gallbladder carcinoma, [47] S. Gourgiotis, H.M. Kocher, L. Solaini, A. Yarollahi, E. Tsiambas, N.S. Salemis,
Rev. Med. Chile 132 (11) (2004 Nov) 1369–1376. Gallbladder cancer, Am. J. Surg. 196 (2008) 252–264.
[37] B. Klump, C.J. Hsieh, S. Dette, K. Holzmann, R. Kiebetalich, M. Jung, U. Sinn, [48] I. Roa, J.C. Araya, M. Villaseca, X. De Aretxabala, P. Riedemann, K. Endoh, et al.,
M. Ortner, R. Porschen, M. Gregor, Promoter methylation of INK4a/ARF as detected Preneoplastic lesions and gallbladder cancer: an estimate of the period required for
in bile-significance for the differential diagnosis in biliary disease, Clin. Canc. Res. 9 progression, Gastroenterology 111 (1996) 232e6.
(5) (2003 May) 1773–1778. [49] I. Roa, J.C. Araya, M. Villaseca, J. Roa, X. de Aretxabala, G. Ibacache, Gallbladder
[38] T. Ueki, A.W. Hsing, Y.T. Gao, B.S. Wang, M.C. Shen, J. Cheng, J. Deng, cancer in high risk area: morphological features and spread patterns, Hepato-
J.F. Fraumeni Jr., A. Rashid, Alterations of p16 and prognosis in biliary tract can- Gastroenterology 46 (1999) 1540e6.
cers from a population-based study in China, Clin. Canc. Res. 10 (5) (2004 Mar 1) [50] A.R. Sepulveda, D. Jones, S. Ogino, W. Samowitz, M.L. Gulley, R. Edwards,
1717–1725. V. Levenson, V.M. Pratt, B. Yang, K. Nafa, L. Yan, P. Vitazka, CpG methylation
[39] J. Behrens, B. Lustig, The Wnt connection to tumorigenesis, Int. J. Dev. Biol. 48 analysis–current status of clinical assays and potential applications in molecular
(5–6) (2004) 477–487. diagnostics: a report of the association for molecular pathology, J. Mol. Diagn. 11
[40] Y. Huang, Q. Du, W. Wu, F. She, Y. Chen, Rescued expression of WIF-1 in gall- (2009) 266–278.
bladder cancer inhibits tumor growth and induces tumor cell apoptosis with altered [51] E.L. Sceusi, D.S. Loose, C.J. Wray, Clinical implications of DNA methylation in
expression of proteins, Mol. Med. Rep. 14 (3) (2016 Sep) 2573–2581. hepatocellular carcinoma, HPB 13 (2011) 369–376.
[41] B. Lin, H. Hong, X. Jiang, C. Li, S. Zhu, N. Tang, X. Wang, F. She, Y. Chen, WNT [52] K. Ghoshal, S. Bai, DNA methyltransferases as targets for cancer therapy, Drugs
inhibitory factor 1 promoter hypermethylation is an early event during gallbladder Today 43 (2007) 395–422.

183

You might also like