Differential Role of microRNAs in The Pathogenesis An - 2016 - Biomedicine - Pha

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Biomedicine & Pharmacotherapy 82 (2016) 509–519

Available online at

ScienceDirect
www.sciencedirect.com

Differential role of microRNAs in the pathogenesis and treatment of


Esophageal cancer
Maryam Hemmatzadeha,b,c,1, Hamed Mohammadia,c,1, Mohammad Karimia,b,c,
Mohammad Hossein Musavishenasa,b,c , Behzad Baradarana,*
a
Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
b
Tabriz University of Medical Sciences, International Branch (Aras), Tabriz, Iran
c
Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran

A R T I C L E I N F O A B S T R A C T

Article history:
Received 20 February 2016 Esophageal cancer (EC) is the most invasive disease associated with inclusive poor prognosis. EC usually
Received in revised form 6 May 2016 is found as either adenocarcinoma (EAC) or squamous cell carcinomas (ESCC). ESCC forms in squamous
Accepted 9 May 2016 cells and highly occurs in the upper third of the esophagus. EAC appears in glandular cells and ordinarily
develops in the lower one third of the esophagus near the stomach. Barrett’s esophagus (BE) is a
Keywords: metaplastic precursor of EAC. There is a persistent need for improving our understanding of the
MicroRNA molecular basis of this disease. MicroRNAs (miRNAs) demonstrate an uncovered class of small, non-
Esophageal cancer coding RNAs that can negatively regulate the protein coding gene, and are associated with approximately
Treatment
all known physiological and pathological processes, especially cancer. MiRNAs can affect cancer
pathogenesis, playing a crucial role as either oncogenes or tumor suppressors. The recent emergence of
observations on the role of miRNAs in cancer and their functions has induced many investigations to
examine their relevance to esophageal cancer. In esophageal cancer, miRNA dysregulation plays a crucial
role in cancer prognosis and in patients’ responsiveness to neo-adjuvant and adjuvant therapies. In this
review, the oncogenic, tumor suppressive, and drug resistance related roles of miRNAs, and their
involvement in the pathogenesis and treatment of esophageal cancer were summarized.
ã 2016 Elsevier Masson SAS. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 510
2. Esophageal cancer subtypes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 510
2.1. Esophageal adenocarcinoma (EAC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 510
2.2. Esophageal squamous cell carcinomas (ESCC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 510
3. MiRNAs in the pathogenesis of EC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 510
3.1. Dysregulation of miRNAs in EAC and its precursor BE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 510
3.2. Dysregulation of miRNAs in ESCC development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 512
4. MiRNAs as biomarkers in esophageal cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 512
5. Oncogenic miRNAs in esophageal cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 513
5.1. MiR-21 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 513
5.2. MiR-10b . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 513
5.3. MiR-17-92 polycistron . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 513
5.4. MiR-200c and miR-221/222 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 513
6. Tumor suppressor miRNAs in esophageal cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 514
6.1. Let-7 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 514
6.2. MiR-375 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 514

* Corresponding author.
E-mail address: Behzad_im@yahoo.com (B. Baradaran).
1
These authors contributed equally to this manuscript.

http://dx.doi.org/10.1016/j.biopha.2016.05.009
0753-3322/ã 2016 Elsevier Masson SAS. All rights reserved.
510 M. Hemmatzadeh et al. / Biomedicine & Pharmacotherapy 82 (2016) 509–519

6.3. MiR-145 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 514


6.4. MiR-34a . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 514
7. Chemoresistance miRNAs in esophageal cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 514
7.1. MiR-148a . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 514
7.2. MiR-141 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 514
7.3. MiR-27a . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 515
7.4. MiR-296 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 515
8. Potential in screening and treatment of EC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 516
9. MiRNAs as therapeutic targets in esophageal cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 516
10. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 517
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 517
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 517
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 517

1. Introduction 2. Esophageal cancer subtypes

Esophageal cancer is the eighth most occurring, and the sixth 2.1. Esophageal adenocarcinoma (EAC)
most fatal cancer worldwide [1]. Histologically, in almost 95% of
cases, the disease is found as ESCC or EAC, with the latter being Esophageal adenocarcinoma (EAC), ordinarily forms in the
approximately 50% more prevalent in the United States but at least lower one third of the esophagus and arises from the Barrett’s
20-times less common in Asian countries [1,2]. The EC occurrence esophagus (BE) [7]. EAC, considered as a consequence of chronic
in the high-risk northern Chinese is more than 100/100000, gastroesophageal reflux, is preceded by the progression of BE, a
whereas in the low-risk western Africa can be 20-fold lower [3]. premalignant condition comprising intestinal metaplasia that
ESCC remains the main subtype of EC, especially in China; in progresses as a sequel of gastroesophageal reflux. Not all BE
contrast, EAC is the most common type in Western countries. The patients progress EAC; nevertheless, in a patient with BE, the risk of
pathogenesis of EC remains unclear. 14–21% of cancers (T1 lesions) EAC is 30 times higher than in the general population [15]. Other
and 38–60% of cancers that invade muscle (T2 lesions) are markers of gastroesophageal reflux disease, such as esophageal
associated with spread to lymph nodes [4,5]. ulcer and frequent utilization of the histamine-H2 blockers, are
The most prevalent risk factor for EAC is chronic gastroesopha- also related to the increased risk but do not seem to be the
geal reflux disease, which triggers inflammation in the distal independent risk factors [16]. Drugs that relax the gastroesopha-
esophagus, resulting in progression of the premalignant lesion geal sphincter and augment the reflux, such as anticholinergic
known as BE or intestinal metaplasia [6]. The development of BE to agents, aminophyllines, and beta-blockers, may contribute to the
EAC, progresses through the established histological changes: development of up to 10 percent of these cancers [17].
intestinal metaplasia (BE) to low-grade dysplasia (LGD) to high
grade dysplasia (HGD) to EAC [7]. There is an insistent need for 2.2. Esophageal squamous cell carcinomas (ESCC)
better understanding of the molecular characteristics of the
disease for the progression of clinically useful biomarkers and Esophageal squamous cell carcinoma arises from the squamous
therapeutic modalities. Biomarkers have been explored based on epithelium of the esophagus and is associated with tobacco
the modifications in genomic DNA, and in the expression of specific consumption [7]. Any factor that causes the chronic irritation and
mRNA or protein molecules, or the metabolites, for many years inflammation of the esophageal mucosa seems to increase the
now [8]. MiRNAs have emerged as a novel class of biomolecules prevalence of squamous-cell carcinoma of the esophagus. Sub-
with significant roles in cellular functions in both healthy and stantial alcohol intake, especially in cooperation with smoking,
diseased cells, and have distinguished potential as biomarkers. greatly increases the risk of squamous-cell carcinoma (but not
Functionally, active miRNAs or mature miRNAs, are 18–22 adenocarcinoma) [18]. In Asia, 90% of EC has the histological type of
nucleotide long, single- stranded RNA molecules with 50 phosphate ESCC. Currently, the most operative treatment is surgical resection.
and 30 hydroxyl groups [2]. Pre-miRNAs are created in the nucleus The 5-year survival rate is 20–30 percent for patients who have
by the act of RNAse III endoribonuclease, Drosha. Another curative surgery and do not progress lymph node metastases;
cytoplasmic RNAse III enzyme, Dicer, is bearing stems from the nevertheless, the survival rate is only 13% for patients with at least
stem-loop structures of pre-miRNAs [9]. The double-stranded 1 lymph node metastasis [19].
RNA-binding proteins DGCR8 (DiGeorge critical region 8) and TRBP
(Trans activating response RNA binding protein), work along with 3. MiRNAs in the pathogenesis of EC
Drosha and Dicer, respectively [2]. The pre-miRNA with a typical 50
phosphate and 2-nucleotide 30 overhang is exported into the 3.1. Dysregulation of miRNAs in EAC and its precursor BE
cytoplasm, through exportin 5 (Exp-5), a Ran-dependent nuclear
transport receptor protein [10]. Mature miRNAs motivate their Most EAC cases are caused by BE, a precursor lesion in which the
function by the multi-protein RNA-induced silencing complex squamous epithelium of the esophagus is substituted by a
(RISC) that is also responsible for the phenomenon of RNA metaplastic columnar epithelium. BE is approximated to be
interference, generated by small interfering RNAs (siRNAs). present in 1–2 percent of the general population and confers a
MiRNAs are loaded on RISC complexes where they are unfold into 30-fold increased risk of progressing EAC. The malignant
two single-stranded, mature miRNAs [11,12]. Consequently, a development of BE normally follows the sequence of metaplasia,
mature miRNA can target hundreds of various mRNAs, and the low-grade dysplasia (LGD), high-grade dysplasia (HGD), and
same mRNA can be targeted by the majority of various miRNAs adenocarcinoma [20].
[13,14] (Fig. 1). In this review, we discussed about the role of Some studies have demonstrated the role of miRNAs in the
various miRNAs and their role in pathogenesis and treatment of disease development from Barrett’s to adenocarcinoma. For
esophageal cancer (Table 1). example, Yang et al. [21] compared the samples from BE patients
M. Hemmatzadeh et al. / Biomedicine & Pharmacotherapy 82 (2016) 509–519 511

Fig. 1. Biogenesis of microRNAs. MicroRNAs are transcribed by RNA polymerase II to long primary-miRNA that can be recognized and cleaved by Drosha in the nucleus. These
miRNA is then processed into 70–120-nucleotide-long precursor RNA (pre-miRNA). By exportin 5, a Ran-dependent nuclear transport receptor protein, the pre-miRNA is
exported from the nucleus to the cytoplasm is further processed by Dicer, to produce a duplex microRNA. The passenger miRNA fragment is degraded, whereas the mature
miRNA molecule binds to an Ago protein and incorporates into the RISC. Finally, mature miRNA causes translational repression and target mRNA degradation. DGCR8:
DiGeorge syndrome critical region gene 8, TRBP: Tandem repeat binding protein, RISC: RNA-induced silencing complex.

with low-grade dysplasia, high-grade dysplasia and EAC. They seem to offer that miRNA could play a critical role in the
were incapable to discover any remarkable differentially expressed development of the disease to later stages.
miRNAs between BE with LGD and the paired normal tissues. Feber et al. [22] compared the expression of 328 human miRNAs
When they examined HGD and EAC tissues, they found 24 miRNAs in 10 EAC, 10 ESCC, 5 BE, 1HGD, and 9 normal squamous epithelium
that revealed the modified expression compared to the normal (NSE) tissues. These studies showed that the miRNA expression
tissue (14 up-regulated in the diseased tissues and 10 down- profiles of BE and EAC were similar. The single HGD specimen had a
regulated in the diseased tissues). Nine of the 14 up-regulated miRNA expression profile similar to EAC samples. About 30
miRNAs (hsa-miR-126, hsa-miR-143, hsa-miR-145, hsa-miR-181a, miRNAs were remarkably altered in EAC tissues compared to NSE,
hsa-miR-181b, hsa-miR-199a, hsa-miR-28, and hsa-miR-30a-5p) like upregulation of miR-21, miR-192, mir-194 and miR-93 and
demonstrated greater expression in EAC compared to HGD; 7 of the down-regulation of miR-203, miR-205, miR-27b, miR-100, miR-
10 down-regulated miRNAs (hsa-miR-149, hsa-miR-203, hsa-miR- 125b, and let-7c. Additionally, it seemed that miR-21, miR-194, and
210, hsa-miR-27b, hsa-miR-513, hsa-miR-617, and hsa-miR-99a) miR-192 were progressively upregulated from NSE to BE and to
revealed lower expression in EAC compared to HGD. These data EAC, offering that miRNA dysfunction is an early incident during
512 M. Hemmatzadeh et al. / Biomedicine & Pharmacotherapy 82 (2016) 509–519

Table 1 ESCC cancers. Ogawa et al. [27] quantified the expression of 73


Oncogenic oncosuppressor and Chemoresistance function of MicroRNA in
miRNAs through quantitative reverse transcriptase polymerase
Esophageal cancer.
chain reaction (qRT-PCR) in 30 ESCC tumor and paired normal
Name Tumor Type Target References tissues. About 21 miRNAs, including miR-21, miR-25, and miR-151,
Oncogenic microRNAs were upregulated at least 2-fold; and 4 miRNAs were down-
miR-196a EAC ANXA1, SPRR2C, S100A, KRT5 [35,82] regulated at least 2-fold (miR-133a, miR-133b, miR-139, and miR-
miR-25 EAC Bim, CDH1 [22]
145) in ESCC. Hong et al. [24] determined a panel of 12 miRNAs,
miR-93 EAC P21 [23]
miR-221/222 EAC P27kip1, CDX2 [55]
variously expressed between ESCC and normal tissues with 9
miR-21 ESCC PDCD4 [45] upregulated (miR-155, miR-100, miR-146, miR-296, miR-10b, miR-
miR-10b ESCC KLF4 [50] 203, miR-483, miR-494 and miR-220) and 3 down-regulated (miR-
miR-31 ESCC PPP2R2A, PDCD4 [47] 143, miR-375 and miR-339) in ESCC. There were also a number of
miR-373 ESCC LATS2 [83]
candidate miRNA studies in ESCC. Tian et al. [28] investigated that
Oncosuppressor microRNAs miR-10b was overexpressed in 95% of ESCC tissues. MiR-17-92
let-7 ESCC HMGA2 [57] cluster was found to be up-regulated in 75% of ESCC samples [29].
miR-34a ESCC NF-kB [69] MiR-31 was overexpressed in 78% of the ESCC tissues [30]. MiR-29c
miR-133a ESCC CD47 [84]
level was remarkably lower in ESCC tumor tissues and cell lines
miR-150 ESCC ERK-5, ZEB-1 [85]
miR-375 ESCC PDK-1, IGF1R [60,61]
compared to normal esophageal epithelia [31]. MiR-210 was
miR-205 ESCC ZEB-2 [86] down-regulated in ESCC and derived cell lines, particularly in
miR-145 ESCC FSCN-1 [66] poorly differentiated carcinomas [32]. In summary, the consis-
miR-29c ESCC CCNE [31] tently overexpressed miRNAs in ESCC, such as miR-21, miR-25,
miR-210 ESCC FGFRL1 [87]
miR-151, and miR-10b, and down-regulated miRNAs such as
Chemoresistance microRNAs miR205, miR-203, miR-145, miR-100, miR-99a, miR-27b, miR-
miR-141 ESCC YAP-1 [75] 125b, miR-133a, miR-133b, miR-143 and miR-375, were introduced
miR-148a ESCC, EAC CDC25B, BCL-2 [88,89] [22].
mir-296 ESCC Cyclin D1, p27, P-gp, Bcl-2, Bax [24,71]
miR-27a ESCC P-gp, Bcl-2, Bax [78,90]
miR-214 ESCC PTEN [91]
4. MiRNAs as biomarkers in esophageal cancer

EAC: Esophageal adenocarcinoma, ESCC: Esophageal squamous cell carcinoma,


In order to utilize miRNAs as the diagnostic tools for esophageal
ANXA1: Annexin A1, CDH1: Cadherin1, PDCD4: Programmed cell death 4, KLF4:
Kruppel-like factor 4, LATS2: Large tumor suppressor homolog 2, PDK-1: cancer, varying levels of expression must be found in malignant
Phosphoinositide-dependent kinase 1, IGF1R: Insulin-like growth factor-1 receptor, cells compared to normal or pre-malignant cells; therefore,
FSCN-1: Fascin homolog 1, FGFRL1: Fibroblast growth factor receptor like- 1, YAP-1: specific miRNAs that are remarkably up-regulated or down-
Yes-associated protein-1, PTEN: Phosphatase and tensin homolog.
regulated between these groups must be determined [33]. MiR-
15b, miR-21, miR-203, miR-486-5p and let-7a, has been utilized for
the malignant development of BE and therefore may become the the forecast of BE progression to EAC; it is likely that these miRNAs
useful biomarkers for malignant development of the BE patients. are involved in the development of adenocarcinoma [34]. MiR-196
Kan et al. [23] have also examined a miRNA signature in Barrett’s has also been discovered to increase progressively from normal
tumorigenesis. In a study of 22 normal, 24 BE and 22 EAC tissue squamous mucosa to LGD and then HGD to adenocarcinoma [35].
samples, they determined 3 miRNAs that were up-regulated and 4 In ESCC, miR-34b and miR-139 are found to be consistently
miRNAs that were down-regulated in BE or EAC compared to the dysregulated, while miR-129 has been determined as a remark-
normal epithelium. The theory that the 3 up-regulated miRNAs able diagnostic factor. MiR-34b can be down-regulated in ESCC
would have an oncogenic function is supported through the cells compared to normal ones [36]. Upregulation of miR-25, miR-
finding that inhibitors of miR-25, miR-93 and 106b reduced the in 151 and miR-424 as well as down-regulation of miR-29c, miR-99a,
vivo carcinogenesis in mice. Interestingly, miR-25, miR-93 and miR-100 and miR-140* are capable to forecast the squamous cell
miR-106b were not remarkably up-regulated in BE relative to NE carcinoma compared to adjacent normal tissues [37]. MiR-200b/c,
tissues, but they were significantly up-regulated in EAC relative to miR-205 and miR-429 in ESCC, are remarkably higher, and their
NE and BE. Again, this demonstrates that there is a progressive fluctuation has been validated. Therefore, they can be not only
participation of these miRNAs in the neoplastic process. In biomarkers for ESCC, but could also be the tumor invasion
summary, a number of miRNAs emerged as critical players in markers [38].
the pathogenesis of EAC and may become the useful biomarker of Recent researches have revealed that tumor derived miRNAs are
EAC and/or BE. MiR-21, miR-192, miR-194 were the most resistant to endogenous ribonuclease activity so it can be present
consistently upregulated miRNAs in EAC, whereas miR-203, in human serum in a significantly stable form [39]. In addition, the
miR-205, miR-100, miR-125b, miR-99a, miR-27b and let-7c were expression level of serum miRNAs is reproducible and consistent
consistently downregulated in EAC. Moreover, for miR-21, among individuals [39,40]. These tumor-derived miRNAs are
miR-192, miR-203, miR-205, and let-7c, there appeared to be a present in the circulating blood at levels sufficient to be
gradual increase or decrease of expression from NSE to BE to measurable as biomarkers for the detection of tumors [39].
EAC [22]. Zhang et al. [19] compared the pooled serum samples by Solexa
sequencing and determined 25 miRNAs atypically expressed
3.2. Dysregulation of miRNAs in ESCC development between the samples of ESCC patients and the normal ones. A
panel of 7 serum miRNAs (miR-10a, miR-22, miR-100, miR-127-3p,
ESCC is the prominent histological subtypes in Far East and all of miR-133a, miR-148b, and miR-223) was determined as a potential
the published studies evaluating miRNAs in ESCC have come from biomarker for ESCC. The serum level of miR-200c can be useful for
Asia [24,25]. Faber et al. [6] found that miR-21, miR-93, and miR- predicting the responses to chemotherapy and the prognosis of
342 were up-regulated whereas miR-203, miR-205, miR-27b, miR- patients with EC who receive neo-adjuvant chemotherapy [41].
100, miR-125b, and let-7c were down-regulated in ESCC. Lee et al. The up regulation of miR-21 and down regulation of miR-375 in
[26] discovered that four miRNAs, let-7d, miR-330, miR-340, and plasma samples of ESCC patients compared to normal subjects,
miR-373, were overexpressed by at least 1.5-fold in each of the 5 offered that the plasma level ratio of miR-21 to miR-375 could
M. Hemmatzadeh et al. / Biomedicine & Pharmacotherapy 82 (2016) 509–519 513

reveal a better diagnostic biomarker than either markers alone. dependent G1/S cell cycle arrest in response to DNA damage [22].
Besides, the same Japanese group also assessed the prognostic Furthermore, miR-10b acts as oncomiR and its oncogenic function
value of miR-21 and miR-375 and reported that the high plasma is at least relatively mediated by KLF4 in ESCCs [50].
level of miR-21 is associated with higher risk of reoccurrence and
poorer survival, whereas the high plasma level of miR-375 is the 5.3. MiR-17-92 polycistron
symptom of better survival [42,43].
MiR-17-92 cluster, also called as oncomir-1, is one of the
5. Oncogenic miRNAs in esophageal cancer oncogenic miRNAs that is found overexpressed in malignant
cancers. The gene is settled at 13q31.3, encodes six mature miRNAs
5.1. MiR-21 including miR-17, miR-18a, miR-19a, miR-20a, miR-19b-1, and
miR-92-1, and has been involved in the oncogenesis of multiple
MiR-21 is the most routinely upregulated miRNA in human malignant cancers [29]. The miR-17-92 cluster was discovered to
cancers that shows the most constant association with both EAC be overexpressed in 21 out of 28 (75%) esophageal cancer samples.
and ESCC progression as well as the prognosis of ESCC. MiR-21 In addition, overexpression of the miR-17-92 cluster could elevate
regulates a plethora of target genes that are involved in cellular the cellular growth in vitro and in vivo, and the suppression of miR-
survival, apoptosis and cell aggressiveness, including a number of 19a, induced apoptosis in vitro and impaired tumor growth in vivo.
tumor-suppressor genes such as tropomyosin-1 (TPM1), PTEN, TNF-a (tumor necrosis factor-a) was verified as a direct target of
maspin, and programmed cell death 4 (PDCD4) [44,45] (Fig. 2a). miR-19a and TNF-a-encoded mRNA includes a 30 UTR element that
Down-regulation of miR-21 can suppress the cell proliferation and is relatively complementary to miR-19a [29]. In addition, miR-92a
induce apoptosis, with identified direct targets such as FASL, TIMP3 was found to regulate the migration and invasion but not apoptosis
and RECK [46]. Moreover, the over-expression of miR-31 and of esophagus squamous carcinoma cell lines in vitro. Furthermore,
miR-21 may be associated with the down-regulation of their miR-92a directly targeted CDH1 and suppressed the expression of
respective tumor suppressor targets PPP2R2A and PDCD4 in ESCC. CDH1 [51].
MiR-31 and miR-21 levels are directly related to the appearance of
ESCC [47]. 5.4. MiR-200c and miR-221/222

5.2. MiR-10b Dysregulation of miR-200c was discovered in several types of


cancer, including esophageal cancer [52]. Studies have revealed a
MiR-10b is overexpressed in various types of cancer [48,49] correlation between miR-200c expression and resistance to
including ESCC. Tian et al. [28] found that the cellular miR-10b anticancer drugs. PI3 K/Akt signaling pathways are disrupted in
expression level is correlated with the cell motility and aggres- majority types of tumor, such as ESCC [53] that affect their
siveness in various human ESCC cell lines. Furthermore, they responsiveness to chemotherapy [54]. Consequently, miR-200c
identified KLF4 (Krüppel-like factor 4), a known tumor suppressor influences the responsiveness to chemotherapy and radiotherapy
gene that has been shown to suppress esophageal cancer cell in ESCC, and patients with high expression of serum miR-200c may
migration and aggression, as a direct target of miR-10b. KLF4 endure higher risk of death; thereby the serum level of miR-200c
regulates the expression level of p21 and mediates the p53- can be useful for anticipating responses to chemotherapy [52]. In

Fig. 2. MicroRNAs in cell proliferation and cell death. MicroRNAs controlled the cell destiny by regulating cell proliferation and cell death pathways with their oncogenic and
tumor suppressive function. (a): Role of OncomiRs in cell death and cell proliferation. (b): Role of Tumor suppressors in cell death and cell proliferation. KLF-4: Krüppel-like
factor-4, CDX2: Caudal-related homeobox 2, IGF1R: Insulin-like growth factor-1 receptor, FSCN1: Fascin-1.
514 M. Hemmatzadeh et al. / Biomedicine & Pharmacotherapy 82 (2016) 509–519

addition, miRNA-221/222 acts as an oncogene in EAC. The raised molecular modifications in ESCC and EAC, offering that miR-145
levels of miR-221/222 have also been observed in EAC. The may show its tumor suppressor function in EC through the
exposure of esophageal cells to bile acids may activate farnesoid X p53-Myc network. However, there has not been any direct
receptor and upregulate the levels of miR-221/222, by decreasing evidence exhibiting that miR-145 prevents c-Myc in EC. On the
the levels of p27Kip1 and promoting the degradation of CDX2. other hand, Kano et al. [66] determined a novel target gene, FSCN1
Consequently, down-regulation of p27Kip1, as a result of the (actin-binding protein, Fascin homolog 1), for miR-145. FSCN1 was
increased expression of miR-221/222, increased the degradation of overexpressed in ESCC tumors compared to normal epithelium,
Cdx2 and cell proliferation during the exposure to bile acid in and FSCN1 overexpression was remarkably connected to the extent
human esophageal epithelial cells. This mechanism might play a of the tumor, lymph node metastasis, and poor prognosis [67].
crucial role in the development of esophageal adenocarcinoma These data offer inhibitions of FSCN1, which may be one of the
[55]. mechanisms for the tumor suppressor function of miR-145 in ESCC.

6. Tumor suppressor miRNAs in esophageal cancer 6.4. MiR-34a

6.1. Let-7 MiR-34a has recently been found to act as a critical tumor
suppressor in the progression of different cancers. Some of the
Let-7 was remarkably, discovered as the second miRNA after genes referring to the cell cycle and apoptosis control including
lin-4 in C. elegans and known as one of the most extensive miRNA CDK4/6, cyclin D1, E2F3, MYCN, SIRT1 and Bcl2 are shown to be
[56]. High mobility group A2 (HMGA2) protein, is a non-histone downregulated by miR-34a [68,69]. Although the most critical
DNA binding factor that is highly expressed during embryogenesis regulator of miR-34a expression is the well-known tumor
and overexpressed in different benign and malignant cancers suppressor p53 [70], bioinformatics analysis demonstrated that
[57,58]. There are several let-7 complementary regions at the there were some potential NF-kB binding sites located in the
30 -UTR of HMGA2 and the role of let-7 is significant in regulating promoter region of miR-34a gene. A novel mechanism of miR-34a
HMGA2 protein expression to prevent the cell proliferation in ESCC regulation in ESCC is that NF-kB can elevate the miR-34a
(Fig. 2b). In addition, HMGA2 is negatively regulated by let-7 at the expression levels by the direct binding to its promoter, and the
post-transcriptional level [57]. Also, the expression of let-7 in wild-type p53 is responsible for NF-kB- mediated miR-34a
esophageal cancer could be utilized to forecast the sensitivity/ transcriptional function. This might show a novel perspective
resistance to cisplatin-based chemotherapy, and studies showed for the roles of miR-34a and NF-kB in esophageal cancer
that the high expression of let-7c is remarkably correlated with the development [69].
clinical response in esophageal cancer. Let-7c expression modu-
lated apoptosis in cisplatin-treated cells, through the down- 7. Chemoresistance miRNAs in esophageal cancer
regulation of IL-6– mediated signaling pathway. Moreover,
phosphorylated STAT3, which is the downstream of IL-6, was 7.1. MiR-148a
induced by cisplatin in esophageal cancer cells. Transfection of let-
7c can directly suppress the cisplatin-activated interleukin IL-6/ MiRNA-148a is one of the drug resistance related miRNAs in EC.
STAT3 pro-survival pathway after the genotoxic chemotherapy in The expression levels of miR-148a in EAC are inversely related to
esophageal cancer cells [59]. the cancer differentiation [71]. In patients with locally advanced
ESCC, the expression of miR-148a is associated with the disease
6.2. MiR-375 recurrence and the tumor-related mortality [37]. Hummel et al.
discovered that miR-148a could improve the response to
The promoter of miRNA-375 is regularly hyper methylated in chemotherapy in the responsive and resistant EC cells [72].
esophageal cancer, and miR-375 negatively regulates the 3- Upregulation of miR-148a could remarkably increase the sensitiv-
phosphoinositide dependent protein kinase-1 (PDK1) in EC [60]. ity of EC cell lines to chemotherapy, represented by a reduction in
In addition, the frequent promoter hyper-methylation of miR-375 the cell viability after 5-fluorouracil (5-FU) treatment. Upregula-
was discovered in ESCC tissues [61]. Moreover, miR-375 could tion of miR-148a can sensitize the chemotherapy-sensitive
interact with the 30 UTR of IGF1R and down-regulate its expression esophageal cancer cell lines to cisplatin (CDDP) and decrease
in vitro. Additionally, miR-375 expression level was negatively the resistance in chemotherapy resistant variants (Fig. 3a). There is
correlated the expression of IGF1R in clinical samples [61]. MiRNA- a tendency toward a better response to 5-FU in the 5-FU-
375 can be restored through the cyclic hydroxamic acid-containing responsive and resistant cells following the miR-148a transfection.
peptide 31, a histone deacetylase inhibitor of ESCC cells. LDHB and The exact implicated mechanisms should be examined by further
AEG-1/MTDH are detected as miR-375-targeted genes, with both experimental and clinical studies [71].
mRNA and protein expression levels of these verified in ESCC
clinical specimens [62]. These data prepare compelling evidence 7.2. MiR-141
supporting that the down-regulation of miR-375, by promoter
hyper-methylation, is one of the molecular mechanisms implicat- MiR-141 is located on chromosome 12 and has a particular
ed in the development of ESCC [22]. pattern of expression in human cancers of epithelial cell type. The
expression of miR-141 is attenuated in Barrett’s epithelium when
6.3. MiR-145 compared with gastric and duodenal epithelia [73]. MiR-141 may
also be implicated in the progression of gastric cancer through its
MiRNA-145 is down-regulated in various cancers [63] and inhibitory effect on cell proliferation [74]. Imanaka et al. discovered
precancerous lesions [64]. Particularly, Sachdeva et al. [65] that miR-141 has a critical regulatory function in the development
demonstrated that c-Myc is a direct target for miR-145. Transcrip- of CDDP-resistance in CDDP-resistant ESCC [75]. MiR-141 is highly
tionally, miR-145 is induced by the tumor suppressor p53. expressed in the CDDP-resistant EC cell lines. When expressed
Moreover, miR-145 provides a direct connection between p53 ectopically in the CDDP-sensitive cell lines, the cell viability after
and c-Myc. The loss of heterogeneity (LOH) of 17p13 (p53 locus) CDDP treatment is remarkably augmented. MiR-141 may directly
and the amplification of 8q24 (Myc locus) are both frequent target the 30 untranslated region (UTR) of YAP1, which plays an
M. Hemmatzadeh et al. / Biomedicine & Pharmacotherapy 82 (2016) 509–519 515

Fig. 3. Effect of microRNAs in sensitivity/resistance to chemotherapy. a) Up-regulation of miR-148a remarkably decreases resistance of EC cell lines to chemotherapy by
targeting CDC25B and BCL-2. Also, up-regulation of miR-141 increases drug resistance through targeting the 30 untranslated region (UTR) of YAP1. YAP-1: Yes-associated
protein 1, BCL-2: B-cell lymphoma 2, P-gp: P-glycoprotein. b) Down-regulation of miR-27a and miR-296 may promote ADR-induced apoptosis through regulation of Bcl-2,
Bax, and p-gp.

important role in the DNA damaging agents-induced apoptosis, augmented accumulation of ADR. Down-regulation of miR-27a can
and thus down-regulates the YAP1 expression (Fig. 3a). Moreover, remarkably attenuate the expression of P-gp, Bcl-2, and the
miR-141 may play a crucial regulatory role in the progression of transcription of the MDR gene 1, as well as up-regulate the
CDDP resistance in ESCC [71,75]. expression of Bax, and confer the drug-induced apoptosis through
enhancing the Bcl-2/Bax ratio, thereby effectively reverse the drug
7.3. MiR-27a resistance of esophageal cancer cells [78] (Fig. 3b).

MiR-27a is highly expressed in cancer cells and may act as an 7.4. MiR-296
oncogene through the regulation of cell survival and angiogenesis
[76]. MiR-27a may repress the cdc2/cyclin B inhibitor and thereby MiR-296 is implicated in many physiological and pathological
facilitate the cancer cell proliferation by arresting cells at G2-M processes, such as carcinogenesis, fetal alcohol syndrome, insulin
[77]. MiR-27a is capable of regulating the cell cycle progression and production as well as insulin secretion [79,80]. Inhibition of
angiogenesis following the drug treatment, and delicately cooper- miR-296 with antagomirs may attenuate angiogenesis in tumor
ate with several other gene regulators to ensure the establishment xenografts in vivo. Additionally, miR-296 reveals low levels of
of drug defense network [78]. expression in the NIH3T3 cells after the UVB irradiation [81],
Down-regulation of miR-27a may confer the sensitivity of both determining that miR-296 may be implicated in DNA damage
P-gp-related and P-gp-non-related drugs on EC cells, and may repair. Reduced expression of miR-296 is capable to differentiate
elevate Adriamycin (ADR)-induced apoptosis, accompanied by long term survivors with the node-positive esophageal cancer
516 M. Hemmatzadeh et al. / Biomedicine & Pharmacotherapy 82 (2016) 509–519

from those dying within 20 months. Down-regulation of miR-296 there is an evidence of aberrant miRNA expression being
may prevent the growth of EC cells in vitro and in vivo through implicated in regulating the expression of multi-drug resistance
regulation of cyclin D1 and p27. Down-regulation of miR-296 can (MDR)-related genes [94]. Furthermore, several miRNAs have
confer sensitivity of drugs on EC cells by attenuating the releasing recently been associated with various esophageal tumor responses
amount of Adriamycin (ADR) through the regulation of P-gp [24]. to therapy (Table 2). For instance, miR-148a expression remarkably
Additionally, down-regulation of miR-296 may promote ADR- improves the sensitivity of esophageal cancer cell lines to cisplatin
induced apoptosis through the regulation of Bcl-2 and Bax. or 5-fluorouracil [72], and miR-141 confers the resistance to
Consequently, miR-296 may play crucial roles in the pathogenesis cisplatin-induced apoptosis by targeting YAP1 [75]. In addition, the
of EC and is considered as a potential target for the intervention of decreased miR-27a and miR-296 expression levels enhance the
this malignancy [71] (Fig. 3b). response to anti-tumor drugs and promote cell apoptosis [24,78].
According to results of microarray analysis, miR-192, miR-194
8. Potential in screening and treatment of EC and miR-622 have been selected, and expressions of these three
miRNAs have been revealed to be remarkably diminished after the
Until now, endoscopic biopsy and histopathological examina- neo-adjuvant therapy, confirming the array profiling data.
tions are the golden standards for high-risk subject screening and Significantly, pre-therapeutic intratumoral expression of both
early detection for EC worldwide. We can only determine 1–2 miR-192 and miR-194 is remarkably related to histopathological
percent early carcinomas and 15–20 percent precancerous lesions response of ESCC to the multimodal therapeutic treatment.
in asymptomatic population aged higher than 35 years in high- Consequently, miR-192 and miR-194 can be considered to be
incidence areas of EC through endoscopic biopsy and histopatho- predictive markers for the therapeutic response in multimodality
logical examinations [92]. Yet, nearly 80% asymptomatic popula- therapy of patients with locally advanced EC [95]. As one of the
tions were in the normal range. Thus, there exists over endoscopic most commonly dysregulated miRNAs in many cancers, miR-21 is
application. remarkably related to the pathological stage of ESCC. The impact of
Additionally, standard sterilizing procedure also restricts the miR-21 on PTEN/AKT signaling pathway has been found to be
wide application of endoscopy in asymptomatic population. In abrogated by the novel organometallic arene Ru (II) drug Rawq01.
addition, the projected cost of mass screening in China for Rawq01 can upregulate the expression of PTEN through the miR-21
detection of cancers is increasing annually. An urgent and critical inhibition and therefore prevent the PI3K-AKT pathway [95].
problem to be solved is to attenuate the blindness in the screening, The in vivo stability of miRNAs has been successfully utilized in
minify the range of endoscopy and promote the detection preclinical models of miRNA-based treatments (either alone or
sensitivity of early carcinoma in high-risk subjects [3]. Easily combined with conventional and/or targeted therapies) [94]. Two
accessible and minimal invasive biomarkers for EC and for other main strategies are prevalently focusing on miRNA expression
diseases are much valuable. The PCR technique for detection and [94,96]: 1) applying oligonucleotides (or virus-based constructs) to
quantification of miRNA [93] in the blood would be one of the block oncogenic miRNA expression or replace the loss of tumor
promising tools of screening individuals for EC and other cancers suppressor miRNAs, 2) regulating miRNA expression through
because of its universal application, suitable management, low cost targeting their transcription/processing. Unluckily, both the issues
and high sensitivity. Therefore, miRNAs acting as oncogenic or of tissue-specific delivery and the cellular uptake of synthetic
performing tumor suppressor activities, are novel biomarkers for oligonucleotides are still major obstacles to the progression of
EC therapies [3]. miRNA targeted therapies. Therefore, various chemical modifica-
tions in oligonucleotides have been explored (i.e. morpholinos,
9. MiRNAs as therapeutic targets in esophageal cancer peptide nucleic acids, cholesterol conjugation, and phosphoro-
thioate backbone modifications) [94,97]. In summary, there are
There have been many innovative studies on miRNAs and EC two feasible approaches for utilizing miRNAs as the cancer
treatment. MiRNAs are exceptional candidates for novel molecular therapeutic factors, either through antisense-mediated inhibition
targeting therapies according to their capability to regulate of oncogenic miRNAs or by ‘replacement’ of under-expressed
multiple genes in molecular pathways. The reason for utilizing tumor suppressive miRNAs with miRNA mimetic or viral vector-
miRNAs as potential anti-cancer drugs is based on two major encoded miRNAs [93].
findings: first, miRNA expression can be dysregulated in cancer; Evident inhibitory types of molecule to miRNA are anti-miRNA
second, targeting miRNA expression may alter the cancer oligonucleotides (AMOs), which may inhibit interactions between
phenotype [33]. In esophageal cancer patients, the response to miRNA and the target mRNAs through competition [98]. Utilizing
chemotherapy (both neo-adjuvant and adjuvant) for similar cancer AMOs is intended to block the expression of an oncogenic miRNA,
phenotypes, varies widely from one patient to another [93], and including miR-17-29 cluster, miR-21, miR-122, miR-155 and miR-

Table 2
Therapeutic function of miRNAs in esophageal cancer.

Therapeutic Function MiRNAs


Improves the sensitivity of esophageal cancer cell to cisplatin or 5-fluorouracil. miR-148a
Confers the resistance to cisplatin-induced apoptosis by targeting YAP-1. miR-141
Down-regulation of miR-27a, enhance the response to anti-tumor drugs and promote cell apoptosis miR-27a
Down-regulation of miR-296, enhance the response to anti-tumor drugs and promote cell apoptosis. miR-296
Predictive markers for multimodality therapy of patients with locally advanced EC. miR-192
Predictive markers for multimodality therapy of patients with locally advanced EC. miR-194
Organometallic arene Ru (II) drug Rawq01 prevent the PI3K-AKT pathway through the miR-21 inhibition. miR-21
Blocked with AMOs. miR-17–29 cluster
Blocked with AMOs. miR-122
Blocked with AMOs. miR-155
Blocked with AMOs. miR-221

AMOs: Anti-miRNA oligonucleotides.


M. Hemmatzadeh et al. / Biomedicine & Pharmacotherapy 82 (2016) 509–519 517

221. In addition, the miR-mask, a sequence with excellent [8] J.A. Jankowski, R.D. Odze, Biomarkers in gastroenterology: between hope and
complementarity to the binding site for an endogenous miRNA hype comes histopathology, Am. J. Gastroenterol. 104 (2009) 1093–1096.
[9] G.M. Borchert, W. Lanier, B.L. Davidson, RNA polymerase III transcribes
in a target gene, can also prevent the access of endogenous miRNA human microRNAs, Nat. Struc. Mol. Biol. 13 (2006) 1097–1101.
to its binding site [99]. Moreover, restoring the endogenous [10] E. Lund, S. Güttinger, A. Calado, J.E. Dahlberg, U. Kutay, Nuclear export of
complement of miRNAs in a cell can be attained by either microRNA precursors, Science 303 (2004) 95–98.
[11] A. Khvorova, A. Reynolds, S.D. Jayasena, Functional siRNAs and miRNAs
introducing a short synthetic duplex RNA molecule that is loaded exhibit strand bias, Cell 115 (2003) 209–216.
into the RISC or by motivating the expression of the stem-loop pre- [12] D.S. Schwarz, G. Hutvágner, T. Du, Z. Xu, N. Aronin, P.D. Zamore, Asymmetry in
miRNA in a viral vector expression system or the miRNA mimetic the assembly of the RNAi enzyme complex, Cell 115 (2003) 199–208.
[13] D.P. Bartel, MicroRNAs: genomics, biogenesis, mechanism, and function, Cell
system, which mimics the endogenous mature miRNA, including 116 (2004) 281–297.
let-7, miR-15 and miR-34 [100]. [14] M. Hemmatzadeh, H. Mohammadi, F. Jadidi-Niaragh, F. Asghari, M. Yousefi,
The role of oncomirs in the pathogenesis and treatment of breast cancer,
Biomed. Pharmacother. 78 (2016) 129–139.
10. Conclusion
[15] E.M. van Soest, J.P. Dieleman, P.D. Siersema, M.C. Sturkenboom, E.J. Kuipers,
Increasing incidence of Barrett’s oesophagus in the general population, Gut
Many comprehensive miRNA expression profiling studies have 54 (2005) 1062–1066.
been appeared in EAC and ESCC tissues. MiRNAs clearly play [16] D.C. Farrow, T.L. Vaughan, C. Sweeney, M.D. Gammon, W.-H. Chow, H.A. Risch,
et al., Gastroesophageal reflux disease, use of H2 receptor antagonists, and
critical roles in the pathogenesis of EAC and ESCC. A number of risk of esophageal and gastric cancer, Cancer Causes Control 11 (2000) 231–
consistently dysregulated miRNAs have been determined in EC, 238.
such as upregulation of miR-21, miR-31, miR-221/222, miR-17-92 [17] J. Lagergren, R. Bergström, H.-O. Adami, O. Nyrén, Association between
medications that relax the lower esophageal sphincter and risk for
polycistron, miR-10b, miR-200c, and miR-373, and down-regula- esophageal adenocarcinoma, Ann. Intern. Med. 133 (2000) 165–175.
tion of miR-375, miR-203, let-7, miR-145, miR-34a, miR-143, miR- [18] L.M. Brown, R. Hoover, D. Silverman, D. Baris, R. Hayes, G.M. Swanson, et al.,
210, and 29c. Consequently, the altered miRNA expression Excess incidence of squamous cell esophageal cancer among US Black men:
role of social class and other risk factors, Am. J. Epidemiol. 153 (2001) 114–
patterns may exert a remarkable effect on the balance between 122.
the tumor suppressors and oncogenes. The field of miRNA [19] C. Zhang, C. Wang, X. Chen, C. Yang, K. Li, J. Wang, et al., Expression profile of
research is very promising in the area of cancer research and microRNAs in serum: a fingerprint for esophageal squamous cell carcinoma,
Clin. Chem. 56 (2010) 1871–1879.
therapy, and the pharmaceutical companies have been trying to
[20] N.J. Shaheen, J.E. Richter, Barrett's oesophagus, Lancet 373 (2009) 850–861.
accelerate the progress of therapeutic miRNA technologies. The [21] H. Yang, J. Gu, K.K. Wang, W. Zhang, J. Xing, Z. Chen, et al., MicroRNA
finding that miRNAs play a critical role in MDR of EC has laid the expression signatures in Barrett's esophagus and esophageal
adenocarcinoma, Clin. Cancer Res. 15 (2009) 5744–5752.
foundation for potential effective clinical management of EC
[22] J. Gu, Y. Wang, X. Wu, MicroRNA in the pathogenesis and prognosis of
cases. The finding of drug resistance-related miRNAs in EC has esophageal cancer, Curr. Pharm. Des. 19 (2013) 1292–1300.
helped to show novel mechanisms in MDR. Continuous expansion [23] T. Kan, F. Sato, T. Ito, N. Matsumura, S. David, Y. Cheng, et al., The miR-106b-25
of determined miRNAs as biomarkers and therapeutic targets may polycistron activated by genomic amplification, functions as an oncogene by
suppressing p21 and Bim, Gastroenterology 136 (2009) 1689–1700.
suggest the fascinating insights into the complexity of gene [24] L. Hong, Y. Han, H. Zhang, M. Li, T. Gong, L. Sun, et al., The prognostic and
expression regulation. In addition, performing research into chemotherapeutic value of miR-296 in esophageal squamous cell carcinoma,
miRNA participation in esophageal carcinogenesis is a rewarding Ann. Surg. 251 (2010) 1056–1063.
[25] S. Kimura, S. Naganuma, D. Susuki, Y. Hirono, A. Yamaguchi, S. Fujieda, et al.,
field, and studying altered miRNA expression patterns can suggest Expression of microRNAs in squamous cell carcinoma of human head and
substantial insights into the pathogenesis of this devastating neck and the esophagus: miR-205 and miR-21 are specific markers for HNSCC
disease. Further work to determine the down-stream targets of and ESCC, Oncol. Rep. 23 (2010) 1625–1633.
[26] K.-H. Lee, Y.-G. Goan, M. Hsiao, C.-H. Lee, S.-H. Jian, J.-T. Lin, et al., MicroRNA-
these miRNAs will improve our understanding of their mecha- 373 (miR-373) post-transcriptionally regulates large tumor suppressor,
nisms of action, which will shed new light on utilizing them homolog 2 (LATS2) and stimulates proliferation in human esophageal cancer,
clinically as biomarkers or therapeutic targets in esophageal Exp. Cell Res. 315 (2009) 2529–2538.
[27] R. Ogawa, H. Ishiguro, Y. Kuwabara, M. Kimura, A. Mitsui, T. Katada, et al.,
cancer. Expression profiling of micro-RNAs in human esophageal squamous cell
carcinoma using RT-PCR, Med. Mol. Morphol. 42 (2009) 102–109.
Conflict of interest [28] Y. Tian, A. Luo, Y. Cai, Q. Su, F. Ding, H. Chen, et al., MicroRNA-10b promotes
migration and invasion through KLF4 in human esophageal cancer cell lines,
J. Biol. Chem. 285 (2010) 7986–7994.
The authors declare no conflict of interest. [29] M. Liu, Z. Wang, S. Yang, W. Zhang, S. He, C. Hu, et al., TNF-a is a novel target of
miR-19a, Int. J. Oncol. 38 (2011) 1013–1022.
Acknowledgments [30] T. Zhang, Q. Wang, D. Zhao, Y. Cui, B. Cao, L. Guo, et al., The oncogenetic role of
microRNA-31 as a potential biomarker in oesophageal squamous cell
carcinoma, Clin. Sci. 121 (2011) 437–447.
None. [31] D.-P. Ding, Z.-L. Chen, X.-H. Zhao, J.-W. Wang, J. Sun, Z. Wang, et al., miR-29c
induces cell cycle arrest in esophageal squamous cell carcinoma by
modulating cyclin E expression, Carcinogenesis 32 (2011) 1025–1032.
References [32] S. Tsuchiya, T. Fujiwara, F. Sato, Y. Shimada, E. Tanaka, Y. Sakai, et al.,
MicroRNA-210 regulates cancer cell proliferation through targeting
[1] M. Hongo, Y. Nagasaki, T. Shoji, Epidemiology of esophageal cancer: orient to fibroblast growth factor receptor-like 1 (FGFRL1), J. Biol. Chem. 286 (2011)
occident. Effects of chronology, geography and ethnicity, J. Gastroenterol. 420–428.
Hepatol. 24 (2009) 729–735. [33] J. Huang, S.Y. Zhang, Y.M. Gao, Y.F. Liu, Y.B. Liu, Z.G. Zhao, et al., MicroRNAs as
[2] S.K. Patnaik, R. Mallick, S. Yendamuri, MicroRNAs and esophageal cancer, J. oncogenes or tumour suppressors in oesophageal cancer: potential
Gastrointestinal Oncol. 1 (2010) 55. biomarkers and therapeutic targets, Cell Prolif. 47 (2014) 277–286.
[3] S.-L. Zhou, L.-D. Wang, Circulating microRNAs: novel biomarkers for [34] A. Bansal, I. Lee, X. Hong, V. Anand, S. Mathur, S. Gaddam, et al., Feasibility of
esophageal cancer, World J. Gastroenterol. 16 (2010) 2348. microRNAs as biomarkers for Barrett's esophagus progression: a pilot cross-
[4] J.-M. Collard, J.-B. Otte, R. Fiasse, P.-F. Laterre, M. De Kock, J. Longueville, et al., sectional phase 2 biomarker study, Am. J. Gastroenterol. 106 (2011) 1055–
Skeletonizing en bloc esophagectomy for cancer, Ann. Surg. 234 (2001) 25. 1063.
[5] J.R. Siewert, H.J. Stein, M. Feith, B.L. Bruecher, H. Bartels, U. Fink, Histologic [35] D.M. Maru, R.R. Singh, C. Hannah, C.T. Albarracin, Y.X. Li, R. Abraham, et al.,
tumor type is an independent prognostic parameter in esophageal cancer: MicroRNA-196a is a potential marker of progression during Barrett's
lessons from more than 1,000 consecutive resections at a single center in the metaplasia-dysplasia-invasive adenocarcinoma sequence in esophagus, Am.
Western world, Ann. Surg. 234 (2001) 360. J. Pathol. 174 (2009) 1940–1948.
[6] A. Feber, L. Xi, J.D. Luketich, A. Pennathur, R.J. Landreneau, M. Wu, et al., [36] L. He, X. He, L.P. Lim, E. De Stanchina, Z. Xuan, Y. Liang, et al., A microRNA
MicroRNA expression profiles of esophageal cancer, J. Thoracic Cardiovasc. component of the p53 tumour suppressor network, Nature 447 (2007) 1130–
Surg. 135 (2008) 255–260. 1134.
[7] S. David, S.J. Meltzer, MicroRNA involvement in esophageal carcinogenesis, [37] R. Hummel, D.J. Hussey, M.Z. Michael, J. Haier, M. Bruewer, N. Senninger, et al.,
Curr. Opin. Pharmacol. 11 (2011) 612–616. MiRNAs and their association with locoregional staging and survival
518 M. Hemmatzadeh et al. / Biomedicine & Pharmacotherapy 82 (2016) 509–519

following surgery for esophageal carcinoma, Ann. Surg. Oncol. 18 (2011) 253– [65] M. Sachdeva, S. Zhu, F. Wu, H. Wu, V. Walia, S. Kumar, et al., p53 represses c-
260. Myc through induction of the tumor suppressor miR-145, Proc. Natl. Acad.
[38] A.K. Shah, N.A. Saunders, A. Barbour, M.M. Hill, Early diagnostic biomarkers Sci. 106 (2009) 3207–3212.
for esophageal adenocarcinoma-the current state of play, Cancer Epidemiol. [66] M. Kano, N. Seki, N. Kikkawa, L. Fujimura, I. Hoshino, Y. Akutsu, et al., miR-145,
Biomarkers Prevention (2013) (cebp. 1415.2012). miR-133a and miR-133b: Tumor-suppressive miRNAs target FSCN1 in
[39] P.S. Mitchell, R.K. Parkin, E.M. Kroh, B.R. Fritz, S.K. Wyman, E.L. Pogosova- esophageal squamous cell carcinoma, Int. J. Cancer 127 (2010) 2804–2814.
Agadjanyan, et al., Circulating microRNAs as stable blood-based markers for [67] Y. Hashimoto, T. Ito, H. Inoue, T. Okumura, E. Tanaka, S. Tsunoda, et al.,
cancer detection, Proc. Natl. Acad. Sci. 105 (2008) 10513–10518. Prognostic significance of fascin overexpression in human esophageal
[40] X. Chen, Y. Ba, L. Ma, X. Cai, Y. Yin, K. Wang, et al., Characterization of squamous cell carcinoma, Clin. Cancer Res. 11 (2005) 2597–2605.
microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and [68] J.S. Wei, Y.K. Song, S. Durinck, Q.-R. Chen, A.T.C. Cheuk, P. Tsang, et al., The
other diseases, Cell Res. 18 (2008) 997–1006. MYCN oncogene is a direct target of miR-34a, Oncogene 27 (2008) 5204–
[41] K. Tanaka, H. Miyata, M. Yamasaki, K. Sugimura, T. Takahashi, Y. Kurokawa, 5213.
et al., Circulating miR-200c levels significantly predict response to [69] J. Li, K. Wang, X. Chen, H. Meng, M. Song, Y. Wang, et al., Transcriptional
chemotherapy and prognosis of patients undergoing neoadjuvant activation of microRNA-34a by NF-kappa B in human esophageal cancer cells,
chemotherapy for esophageal cancer, Ann. Surg. Oncol. 20 (2013) 607–615. BMC Mol. Biol. 13 (2012) 4.
[42] S. Komatsu, D. Ichikawa, H. Takeshita, M. Tsujiura, R. Morimura, H. Nagata, [70] V. Tarasov, P. Jung, B. Verdoodt, D. Lodygin, A. Epanchintsev, A. Menssen, et al.,
et al., Circulating microRNAs in plasma of patients with oesophageal Differential regulation of microRNAs by p53 revealed by massively parallel
squamous cell carcinoma, Br. J. Cancer 105 (2011) 104–111. sequencing: miR-34a is a p53 target that induces apoptosis and G1-arrest,
[43] S. Komatsu, D. Ichikawa, H. Takeshita, H. Konishi, H. Nagata, S. Hirajima, et al., Cell cycle. 6 (2007) 1586–1593.
Prognostic impact of circulating miR-21 and miR-375 in plasma of patients [71] L. Hong, Y. Han, Q. Lu, H. Zhang, Q. Zhao, K. Wu, et al., Drug resistance-related
with esophageal squamous cell carcinoma, Expert Opin. Biol. Ther. 12 (2012) microRNAs in esophageal cancer, Expert Opin. Biol. Ther. 12 (2012) 1487–
S53–S59. 1494.
[44] S.D. Selcuklu, M.A. Donoghue, C. Spillane, miR-21 as a key regulator of [72] R. Hummel, D.I. Watson, C. Smith, J. Kist, M.Z. Michael, J. Haier, et al., Mir-148a
oncogenic processes, Biochem. Soc. Trans. 37 (2009) 918. improves response to chemotherapy in sensitive and resistant oesophageal
[45] C. Kestens, P.D. Siersema, J.W. van Baal, Current understanding of the adenocarcinoma and squamous cell carcinoma cells, J. Gastrointest. Surg. 15
functional roles of aberrantly expressed microRNAs in esophageal cancer, (2011) 429–438.
World J. Gastroenterol. 22 (2016) 1. [73] J.C. Gonzales, L.M. Fink, O.B. Goodman, J.T. Symanowski, N.J. Vogelzang, D.C.
[46] N. Wang, C.-q. Zhang, J.-h. He, X.-f. Duan, Y.-y. Wang, X. Ji, et al., MiR-21 down- Ward, Comparison of circulating MicroRNA 141 to circulating tumor cells,
regulation suppresses cell growth, invasion and induces cell apoptosis by lactate dehydrogenase, and prostate-specific antigen for determining
targeting FASL, TIMP3, and RECK genes in esophageal carcinoma, Dig. Dis. Sci. treatment response in patients with metastatic prostate cancer, Clin.
58 (2013) 1863–1870. Genitourin. Cancer 9 (2011) 39–45.
[47] H. Alder, C. Taccioli, H. Chen, Y. Jiang, K.J. Smalley, P. Fadda, et al., [74] F.Y. Agaoglu, M. Kovancilar, Y. Dizdar, E. Darendeliler, S. Holdenrieder, N.
Dysregulation of miR-31 and miR-21 induced by zinc deficiency promotes Dalay, et al., Investigation of miR-21, miR-141, and miR-221 in blood
esophageal cancer, Carcinogenesis (2012). circulation of patients with prostate cancer, Tumor Biol. 32 (2011) 583–588.
[48] T. Sasayama, M. Nishihara, T. Kondoh, K. Hosoda, E. Kohmura, MicroRNA-10b [75] Y. Imanaka, S. Tsuchiya, F. Sato, Y. Shimada, K. Shimizu, G. Tsujimoto,
is overexpressed in malignant glioma and associated with tumor invasive MicroRNA-141 confers resistance to cisplatin-induced apoptosis by targeting
factors, uPAR and RhoC, Int. J. Cancer 125 (2009) 1407–1413. YAP1 in human esophageal squamous cell carcinoma, J. Hum. Genet. 56
[49] E.O.D.A. Lal, MicroRNAs and their target gene networks in breast cancer, (2011) 270–276.
Breast Cancer Res. 12 (2010) 201. [76] C. Urbich, D. Kaluza, T. Frömel, A. Knau, K. Bennewitz, R.A. Boon, et al.,
[50] H. Xu, Y. Yao, F. Meng, X. Qian, X. Jiang, X. Li, et al., Predictive value of serum MicroRNA-27a/b controls endothelial cell repulsion and angiogenesis by
miR-10b, miR-29c, and miR-205 as promising biomarkers in esophageal targeting semaphorin 6A, Blood 119 (2012) 1607–1616.
squamous cell carcinoma screening, Medicine (Baltimore). 94 (2015) e1558. [77] C. Spruck, miR-27a regulation of SCFFbw7 in cell division control and cancer,
[51] Z.-l. Chen, X.-h. Zhao, J.-w. Wang, B.-z. Li, Z. Wang, J. Sun, et al., microRNA-92a Cell Cycle 10 (2011) 3232–3232.
promotes lymph node metastasis of human esophageal squamous cell [78] H. Zhang, M. Li, Y. Han, L. Hong, T. Gong, L. Sun, et al., Down-regulation of miR-
carcinoma via E-cadherin, J. Biol. Chem. 286 (2011) 10725–10734. 27a might reverse multidrug resistance of esophageal squamous cell
[52] H. Yu, B. Duan, L. Jiang, M. Lin, H. Sheng, J. Huang, et al., Serum miR-200c and carcinoma, Dig. Dis. Sci. 55 (2010) 2545–2551.
clinical outcome of patients with advanced esophageal squamous cancer [79] E. Langenkamp, P.J. Zwiers, H.E. Moorlag, W.P. Leenders, B.S. Croix, G.
receiving platinum-based chemotherapy, Am. J. Trans. Res. 6 (2014) 71. Molema, Vascular endothelial growth factor receptor 2 inhibition in-vivo
[53] J.Á.F. Vara, E. Casado, J. de Castro, P. Cejas, C. Belda-Iniesta, M. González- affects tumor vasculature in a tumor type-dependent way and
Barón, PI3K/Akt signalling pathway and cancer, Cancer Treatment Rev. 30 downregulates vascular endothelial growth factor receptor 2 protein without
(2004) 193–204. a prominent role for miR-296, Anticancer Drugs 23 (2012) 161–172.
[54] H.A. Burris III, Overcoming acquired resistance to anticancer therapy: focus [80] J.E. Robson, S.A. Eaton, P. Underhill, D. Williams, J. Peters. MicroRNAs 296 and
on the PI3K/AKT/mTOR pathway, Cancer Chemother. Pharmacol. 71 (2013) 298 are imprinted and part of the GNAS/Gnas cluster and miR-296 targets
829–842. IKBKE and Tmed9, RNA 18 (2012) 135–144.
[55] J. Matsuzaki, H. Suzuki, H. Tsugawa, M. Watanabe, S. Hossain, E. Arai, et al., [81] S.C. Cazanave, J.L. Mott, N.A. Elmi, S.F. Bronk, H.C. Masuoka, M.R. Charlton,
Bile acids increase levels of microRNAs 221 and 222, leading to degradation of et al., A role for miR-296 in the regulation of lipoapoptosis by targeting
CDX2 during esophageal carcinogenesis, Gastroenterology 145 (2013) 1300– PUMA, J. Lipid Res. 52 (2011) 1517–1525.
1311. [82] E. Stanitz, K. Juhasz, K. Gombos, K. GÅcze, C. Toth, I. Kiss, Alteration of miRNA
[56] B.J. Reinhart, F.J. Slack, M. Basson, A.E. Pasquinelli, J.C. Bettinger, A.E. Rougvie, expression correlates with lifestyle, social and environmental determinants
et al., The 21-nucleotide let-7 RNA regulates developmental timing in in esophageal carcinoma, Anticancer Res. 35 (2015) 1091–1097.
Caenorhabditis elegans, Nature 403 (2000) 901–906. [83] K. Harada, Y. Baba, T. Ishimoto, H. Shigaki, K. Kosumi, N. Yoshida, et al., The
[57] Q. Liu, G.-d. Lv, X. Qin, Y.-h. Gen, S.-t. Zheng, T. Liu, et al., Role of microRNA let- role of microRNA in esophageal squamous cell carcinoma, J. Gastroenterol.
7 and effect to HMGA2 in esophageal squamous cell carcinoma, Mol. Biol. (2016) 1–11.
Rep. 39 (2012) 1239–1246. [84] S. Suzuki, T. Yokobori, N. Tanaka, M. Sakai, A. Sano, T. Inose, et al., CD47
[58] F. Asghari, N. Haghnavaz, B. Baradaran, M. Hemmatzadeh, T. Kazemi, Tumor expression regulated by the miR-133a tumor suppressor is a novel prognostic
suppressor microRNAs: targeted molecules and signaling pathways in breast marker in esophageal squamous cell carcinoma, Oncol. Rep. 28 (2012) 465–
cancer, Biomed. Pharmacother. 81 (2016) 305–317. 472.
[59] K. Sugimura, H. Miyata, K. Tanaka, R. Hamano, T. Takahashi, Y. Kurokawa, [85] T. Yokobori, S. Suzuki, N. Tanaka, T. Inose, M. Sohda, A. Sano, et al., MiR-150 is
et al., Let-7 expression is a significant determinant of response to associated with poor prognosis in esophageal squamous cell carcinoma via
chemotherapy through the regulation of IL-6/STAT3 pathway in esophageal targeting the EMT inducer ZEB1, Cancer Sci. 104 (2013) 48–54.
squamous cell carcinoma, Clin. Cancer Res. 18 (2012) 5144–5153. [86] K. Matsushima, H. Isomoto, N. Yamaguchi, N. Inoue, H. Machida, T. Nakayama,
[60] X. Li, R. Lin, J. Li. Epigenetic silencing of microRNA-375 regulates PDK1 et al., MiRNA-205 modulates cellular invasion and migration via regulating
expression in esophageal cancer, Dig. Dis. Sci. 56 (2011) 2849–2856. zinc finger E-box binding homeobox 2 expression in esophageal squamous
[61] K.L. Kong, D.L.W. Kwong, T.H.-M. Chan, S.Y.-K. Law, L. Chen, Y. Li, et al., cell carcinoma cells, J. Transl. Med. 9 (2011) (10.1186).
MicroRNA-375 inhibits tumour growth and metastasis in oesophageal [87] K. Dang, K.A. Myers, The role of hypoxia-Induced miR-210 in cancer
squamous cell carcinoma through repressing insulin-like growth factor 1 progression, Int. J. Mol. Sci. 16 (2015) 6353–6372.
receptor, Gut (2011) gutjnl-2011-300178. [88] S.-T. Liffers, J.B. Munding, M. Vogt, J.D. Kuhlmann, B. Verdoodt, S. Nambiar,
[62] Y. Isozaki, I. Hoshino, N. Nohata, T. Kinoshita, Y. Akutsu, N. Hanari, et al., et al., MicroRNA-148a is down-regulated in human pancreatic ductal
Identification of novel molecular targets regulated by tumor suppressive adenocarcinomas and regulates cell survival by targeting CDC25B, Lab.
miR-375 induced by histone acetylation in esophageal squamous cell Invest. 91 (2011) 1472–1479.
carcinoma, Int. J. Oncol. 41 (2012) 985–994. [89] H. Zhang, Y. Li, Q. Huang, X. Ren, H. Hu, H. Sheng, et al., MiR-148a promotes
[63] M. Sachdeva, Y.-Y. Mo, miR-145-mediated suppression of cell growth, apoptosis by targeting Bcl-2 in colorectal cancer, Cell Death Differ. 18 (2011)
invasion and metastasis, Am. J. Trans. Res. 2 (2010) 170. 1702–1710.
[64] L.F. Sempere, M. Christensen, A. Silahtaroglu, M. Bak, C.V. Heath, G. Schwartz, [90] D.D. Feng, H. Zhang, P. Zhang, Y.S. Zheng, X.J. Zhang, B.W. Han, et al., Down-
et al., Altered MicroRNA expression confined to specific epithelial cell regulated miR-331–5p and miR-27a are associated with chemotherapy
subpopulations in breast cancer, Cancer Res. 67 (2007) 11612–11620. resistance and relapse in leukaemia, J. Cell. Mol. Med. 15 (2011) 2164–2175.
M. Hemmatzadeh et al. / Biomedicine & Pharmacotherapy 82 (2016) 509–519 519

[91] Y.-S. Wang, Y.-H. Wang, H.-P. Xia, S.-W. Zhou, G. Schmid-Bindert, C.-C. Zhou, indicates a high potential of miR-192 in prediction of multimodality therapy
MicroRNA-214 regulates the acquired resistance to gefitinib via the PTEN/ response, Int. J. Cancer 133 (2013) 2454–2463.
AKT pathway in EGFR-mutant cell lines, Asian Pac. J. Cancer Prev. 13 (2012) [96] J.T. Mendell, E.N. Olson, MicroRNAs in stress signaling and human disease,
255–260. Cell 148 (2012) 1172–1187.
[92] C. Chen, D.A. Ridzon, A.J. Broomer, Z. Zhou, D.H. Lee, J.T. Nguyen, et al., Real- [97] S. Sandhu, R. Garzon, Potential applications of microRNAs in cancer diagnosis,
time quantification of microRNAs by stem–loop RT–PCR, Nucleic Acids Res. prognosis, and treatment, Seminars in oncology, Elsevier, 2011, pp. 781–787.
33 (2005) e179–e179. [98] K. Lennox, M. Behlke, Chemical modification and design of anti-miRNA
[93] M. Fassan, R. Baffa, A. Kiss, G. Zaninotto, M. Rugge, MicroRNA dysregulation in oligonucleotides, Gene Ther. 18 (2011) 1111–1120.
esophageal neoplasia: the biological rationale for novel therapeutic options, [99] S. Nana-Sinkam, C. Croce, Clinical applications for microRNAs in cancer, Clin.
Curr. Pharm. Des. 19 (2013) 1236–1241. Pharmacol. Ther. 93 (2013) 98–104.
[94] R. Garzon, G. Marcucci, C.M. Croce, Targeting microRNAs in cancer: rationale, [100] W.C. Cho, MicroRNAs as therapeutic targets and their potential applications
strategies and challenges, Nat. Rev. Drug Discov. 9 (2010) 775–789. in cancer therapy, Expert Opin. Ther. Targets 16 (2012) 747–759.
[95] M. Odenthal, E. Bollschweiler, P. Grimminger, W. Schröder, J. Brabender, U.
Drebber, et al., MicroRNA profiling in locally advanced esophageal cancer

You might also like