You are on page 1of 37

MINI-REVIEW

Progress and Prospects of Circular RNAs in Hepatocellular Carcinoma: Novel


insights into their function

Ji Hua;b;1, Peng Lic;1, Yang Songd;1, Yun-xuan Gee, Xiao-ming Menga;b, Cheng
Huanga;b, Jun Lia;b, Tao Xua;b*
a
School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products,
Anhui Medical University, Hefei 230032, China
b
Institute for Liver Diseases of Anhui Medical University, Anhui Medical University,
Hefei 230032, China
c
Department of Medical, The First Affiliated Hospital of Anhui Medical University,
Hefei 230032, China
d
Department of Pain treatment, The First Affiliated Hospital of Anhui Medical
University, Hefei 230032, China
e
Beijing Institute of Radiation Medicine, Academy of Military Medical Sciences,
Beijing 100850, China
1
They contribute equally to this work.

* Corresponding author
Tao Xu, PhD
School of Pharmacy,
Anhui Medical University,
81 Meishan Road,
Hefei, Anhui Province, 230032,
China.
Tel./fax: +86 551 63699299
E-mail: xutao@ahmu.edu.cn; ahmuxutao@163.com

This article has been accepted for publication and undergone full peer review but has not been
through the copyediting, typesetting, pagination and proofreading process, which may lead to
differences between this version and the Version of Record. Please cite this article as doi:
[10.1002/jcp.26154]

Received 21 June 2017; Revised 10 August 2017; Accepted 11 August 2017


Journal of Cellular Physiology
This article is protected by copyright. All rights reserved
DOI 10.1002/jcp.26154

This article is protected by copyright. All rights reserved


Abbreviations
Hepatocellular carcinoma (HCC), Circular RNAs (circRNAs) , hepatitis B virus (HBV), hepatitis
C virus (HCV), Long non-coding RNA (lncRNA), MicroRNA (miRNA), Atherosclerotic vascular
disease (ASVD), Alzheimer's disease (AD), Gastric cancer (GC), CiRNAs(circular intronic
RNAs), EcircRNAs (circular exonic RNAs), EiciRNAs (exon-intron circRNAs), Laryngeal
squamous cell carcinoma (LSCC), CDR1 (cerebellar degeneration-related protein 1), eIF4F
(cap-dependent translation regulation complex), eIF4G3 (initiation factor 4 gamma 3), Matrix
metallopeptidases (MMPs), PRDM2 (PR/SET Domain 2), VEGF (Vascular endothelial growth
factor), PTEN (Phosphatase and tensin homolog), Liver regeneration (LR), ER-stress
(Endoplasmic reticulum stress), Small cell lung cancer (SCLC), Alpha-fetoprotein (AFP),
Microvascular invasion (MVI), Receiver-operating characteristic (ROC), Area under the ROC
curve (AUC), Tumor-node-metastasis (TNM) stage, Barcelona Clinic Liver Cancer (BCLC) stage,
Nanoparticles co-loaded with microRNA-375 (NPC/miR-375), Extracellular vesicles (EV)

This article is protected by copyright. All rights reserved


Abstract
Hepatocellular carcinoma (HCC) is one of the most predominant subjects of liver
malignancies, which arouses global concern in the recent years. Advanced studies

have found that Circular RNAs (circRNAs) are differentially expressed in HCC, with
its regulatory capacity in HCC pathogenesis and metastasis. However, the underlying
mechanism remains largely unknown. In this review, we summarized the functions

and mechanisms of those aberrantly expressed circRNAs in HCC tissues. We hope to


enlighten more comprehensive studies on the detailed mechanisms of circRNAs and

explore their potential values in clinic applications. It revealed that hsa_circ_0004018


can be used as a potential biomarker in HCC diagnosis, with its superior sensitivity to
alpha-fetoprotein (AFP). Notably, the correlation of circRNA abundance in the

proliferation of liver regeneration (LR) has recently been clarified and different
circRNA profiles served as candidates for nonalcoholic steatohepatitis (NASH)
diagnosis also be discussed. Therefore, the improved understanding of circRNAs in

HCC pathogenesis and metastasis proposed a novel basis for the early diagnosis in
HCC patients, which provides a useful resource to explore the pathogenesis of HCC.
This article is protected by copyright. All rights reserved

Keywords: Circular RNAs; HCC; microRNA; biomarker; liver

This article is protected by copyright. All rights reserved


Introduction

HCC is the most common malignancy of liver and causes a major health problem
for a long time (Jemal et al., 2011). Clinical treatments, such as liver transplantation
and resection represent the most curative therapy for HCC patients at an early stage.
However, the prognosis after curative therapy for HCC always remained
unsatisfactory because of a high incidence of postoperative recurrence rate (Poon,
2009; Zimmerman et al., 2008). Indeed, HCC is the second cause of cancer-related
death all over the world, whether in rich developed countries (Qatar) or poor
developing countries (Congo) (Llovet et al., 2016). Furthermore, HCC has ranked to
the highest rates due to cancer in Asia and Middle Africa and the average age of death
is much lower than that cardiovascular diseases for decades. Interestingly, the
occurrence of HCC was more frequent among males (Jemal et al., 2011). Therefore,
HCC is becoming a severe social problem affecting human health. Moreover, the
major risk factors of HCC include hepatitis C (HCV) and B virus (HBV) infection,
toxins, chronic alcohol abuse, nonalcoholic fatty liver disease and etc. (Levrero, 2006).
Despite new advanced therapeutic were continuously carried out in the last few years,
the survival rate of HCC is still difficult to improve. Whats more, increasing
researches showed the inter-relevant between various oncogenes or anti-oncogenes
and HCC metastasis, which improved new therapeutic approaches to understand the
exact mechanisms of HCC (Yu et al., 2016).
Accumulating evidence showed that non-coding RNAs, such as long non-coding
RNA (lncRNA) and microRNA (miRNA), playing vital regulatory roles in the cellular
biological and physiological process (Araldi and Suarez, 2016; Guttman and Rinn,
2012; Sabin et al., 2013). Of note, circRNAs, a class of non-coding RNAs, are
recognized as a novel profile of endogenous noncoding RNA, which are normally
produced by scrambling of exons at the time of splicing (Rong et al., 2017). Differs from
linear RNAs terminated with 5' caps and 3' tails, circRNAs form covalently closed
loop structures with neither poly-adenylated tail nor 5' to 3' polarity(Cortes-Lopez and
Miura, 2016; Ebbesen et al., 2016; Li et al., 2016a). Moreover, circRNAs are
extensively expressed in tissues, a subgroup displays conservations and specific
developmental-stage patterns across different species (Fischer and Leung, 2017;
Shang et al., 2016). CircRNAs are renowned for viruses, viroid and tetrahymena for
several decades, but there are only a small number of circRNAs discovered in
mammalian at the present. Besides, further research suggested circRNAs were
associated with various diseases, including cancer (Zhao and Shen, 2015). Therefore,
circRNAs may probably be utilized as predictive biomarkers for cancer diagnosis
(Shang et al., 2016). Recent studies also demonstrated that circRNAs played
significant roles in the progression of cerebrovascular and neurological diseases, such
as atherosclerotic vascular disease (ASVD) and Alzheimer's disease (AD) (Holdt et al.,
2016; Lukiw, 2013; Zhao et al., 2016). Additionally, hsa_circ_0000190 expression
was down-regulated in gastric cancer (GC) tissues and plasma samples in GC patients
(Chen et al., 2017b). Meanwhile, hsa_circ_0001649 and hsa_circ_0005075 were

This article is protected by copyright. All rights reserved


confirmed to be new biomarkers of HCC (Qin et al., 2016; Shang et al., 2016).
Nevertheless, the association between circRNAs and HCC remains unclear (Chen et
al., 2017b). CircRNA formed by back-splicing was showed in Figure1.
Based on the previous findings, there is no doubt that circRNAs are involved in
the pathogenesis of HCC and further researches in this area will certainly make great
contributions to the treatment of this disease. In this review, we discuss new
perspectives of circRNAs in HCC, including its therapeutic function and clinic
potential, combining with the informed researches of miRNA. The goal of this review
is to summarize the emerging evidences and propose a new idea to explain how HCC
is coordinated by circRNAs.

Overview of circRNA

CircRNAs were first observed in cytoplasm of eukaryotic cell in 1979 through an


electron microscope and later discovered in higher plants as pathogenic infectious
RNA molecules (Hsu and Coca-Prados, 1979). It is acknowledged that circRNAs are
enriched in cerebellum with a high expression during neuronal differentiation in
mammalian brains. (Hanan et al., 2016; Rybak-Wolf et al., 2015). The expressions of
several circRNAs are higher than those of their canonical linear transcripts of the
same genes, existing in many types of extracellular body fluids, such as urine, saliva,
and blood (Yao et al., 2017a). As a large family of RNA molecules, it is clear that
messenger RNA (mRNA) is transcribed by a single strand of template DNA, conveys
genetic information from DNA to the ribosome and directs the protein synthesis.
However, the following transcription of eukaryotic transcripts mRNA is known as
mRNA precursors (pre-mRNA), which can generate canonical splicing to a linear
RNA or produce a back-splicing circRNA, when the mRNA contains an apparent
backsplice sequence. (Manning and Cooper, 2017) Additionlly, substantial studies
revealed that the biogenesis of circRNAs via back-splicing can be classified into three
types: ciRNAs (circular intronic RNAs), ecircRNAs (circular exonic RNAs) and
EIciRNAs (exon-intron circRNAs). More narrowly, ciRNA is biogenesis from
splicing reaction, containing a C-rich element and a GU-rich element to form a stable
ciRNA, ecircRNA is widespread with exon skipping in human vein umbilical
endothelial cells, while EIciRNA is circularized with retained-intron via base-pairing.
(Li et al., 2017a; Qu et al., 2015a). Different types of circRNAs may perform different
properties. Both ciRNAs and eiciRNAs were combined with the polymerase II
elongation to enhance their expression of corresponding parent genes in the nucleus
(Zhang et al., 2013). However, ecircRNAs were identified as miRNA decoys by
competing miRNA bindings, following the function of regulating miRNA effects on
their coding RNA targets and competing endogenous RNA activities (Li et al., 2017a;
Li et al., 2015; Li et al., 2017c). Recently, circRNAs were refined by RNA-seq-based
studies as one of RNA species with high relevance for molecular oncology and
biology (Bonizzato et al., 2016; Fischer and Leung, 2017). Approximately, over
20,000 different circRNA species were discovered in human and mouse tissues, as

This article is protected by copyright. All rights reserved


well as 700 in Caenorhabditis elegans (Ivanov et al., 2015; Memczak et al., 2013).
Typically, the account of circular isoforms took up 510% of the overall transcripts of
their corresponding coding gene, but certain circRNAs rose up to 200 times higher
than their linear counterparts (Jeck et al., 2013; Salzman et al., 2013).
CircRNAs are widely expressed in mammals, displaying various cell-type
specific expressions (Caiment et al., 2015; Memczak et al., 2013). Of note, circRNAs
are closely relevant to many diseases. For example, the abundance of certain
circRNAs are associated with GC (Chen et al., 2017a). hsa_circ_002059 was reported
as a potential biomarker in the diagnosis of GC (Shang et al., 2016). Ghosal et al.
found 105 diseases in 174 varieties were connected with circular RNAs, including
different carcinomas and neurodegenerative diseases (Wang et al., 2015). Additionally,
upregulated hsa_circRNA_100855 and downregulated hsa_circRNA_104912 were
stably correlated with the progression of Laryngeal squamous cell carcinoma (LSCC)
(Xuan et al., 2016). From what has been discussed above, the results confirmed
circRNAs may have significant function in human diseases, especially in cancer.
Moreover, it demonstrated circRNAs could regulate the proliferation and survival of
cancer cells. To date, the validated function of circRNAs are the tip of the iceberg and
thousands of circRNAs were found in different tissues and cells.(Zhu et al., 2017b).
As is well-known, circRNAs can function as miRNA sponges, intermediating in
RNA processing reactions or modulating in viral and viroid replications (Kulcheski et
al., 2016). However, the universal cognition of circRNAs was turned from being
recognized as a rare curiosity to central regulatory roles in RNA metabolism.
Amounting evidences demonstrated circRNAs played a crucial role in different stages
of biological procession, including cell development, proliferation, metastasis, fate
decision, migration and invasion (He et al., 2017). Present studies also suggested
circRNAs have potential values in clinical applications of human cancer, special focus
on HCC (Dong et al., 2017).

Overview of HCC

HCC ranked to the fifth most prevalent diagnosed cancer and the third leading
cause of lethal malignancy worldwide (Singal and El-Serag, 2015). It was estimated
that primary HCC or malignant hepatoma represent around 90% in primary liver
cancers. Statistics also showed approximately 850,000 people are recently diagnosed
with HCC and the disease accounts at least 700,000 deaths annually around the world.
It was surprised that nearly half of these cases and deaths took place in China,
resulting in 55% patients from Chinaconsequently became the area with highest
prevalence of HCC (Llovet et al., 2016; Shang et al., 2016). Epidemiologically, HCC
mainly occurred in Africa, North America, Western Europe and Asia, with various
causes including the prevalence of HBV and HCV infections, alcohol, smoking,
dietary exposure to aflatoxins, obesity, and diabetes (Herceg and Paliwal, 2011;
Lozano et al., 2012). Besides, liver cirrhosis and aflatoxin are important risk factors
devoted to the complicated pathological process of HCC. Of note, the tumor-related

This article is protected by copyright. All rights reserved


cells of HCC usually metastasize through blood or lymphatic in the process. Once
tumor-related cells invade the body fluids, it is difficult to control the malignant
metastasis (Xiong et al., 2017). Regional and systemic metastases take on the major
reasons for the unsatisfactory prognosis of HCC patients (Chung and Kemeny, 2005).
Additionally, the properties of HCC metastasis are not only the important pathological
factors with high recurrence rate and poor long-term outcome, but also the essential
pathological basis for developing individualized regimens of controlling metastasis
and recurrence. Novel studies reported the p53 tumor suppressor was linked to HCC
metastasis, with its functions to enhance the transformation of adjacent epithelial cells
into HCC tissues (Levine and Oren, 2009; Wahid et al., 2017). Increasing evidence
suggested that many target proteins were widely existed in mammalian genomes,
playing crucial roles in cell biology. Until now, many tumor suppressor genes have
been implemented in diversely pathological processes in HCC metastasis (Sun et al.,
2017; Tong et al., 2017). However, it is still difficult to elucidate the underlying
molecular mechanisms for HCC metastasis, which is critical for therapeutic targets on
HCC (Yao et al., 2017a). In the mechanism research, the cancer-related genes and
cellular pathways during the initiation and development of HCC have been discussed
for many years, such as the RAF/MEK/ERK pathway is constitutively activated,
suggesting a possible role for many pathways in HCC tumorigenesis. However, there is
still no effective means of preventions or treatments for HCC (Hwang et al., 2004).
Accordingly, it is very important to investigate the molecular mechanisms during the
proliferation, apoptosis and invasion of HCC, making breakthrough progress of
diagnostic and therapeutic strategies. Due to the rapidly development of advanced
microarray technology, it seems easier to emulate the general genetic alterations in the
progression of diseases, which may provide neoteric identification of gene targets for
diagnosis and prognosis of tumors (Mou et al., 2017).
Hitherto, the prevailing treatment of HCC includes liver transplantation,
interventional chemotherapy, local ablative therapy, surgical resection, sorafenib and
radiation (Kuo et al., 2017; Xiong et al., 2017). However, diagnosis in the early stage
of HCC remains an obstacle to overcome and the advanced development of
noninvasive diagnostic tools is facing a major challenge. While, newly discoveries
identified the distinct non-coding RNAs were enriched in HCC exosomes, which
strongly encouraged the idea of using these molecules as novel HCC biomarkers
(Takahashi et al., 2014). With the rapidly improvement of treatment strategies and
diagnostic approaches, people with HCC will be detected at an early stage and accept
immediate radical surgeries with favorable prognosis in the near future rather than
missing the best operation opportunities.

Roles of circRNAs in HCC

Multiple lines of evidences indicated many non-coding RNAs were correlated


with liver diseases. Furthermore, the correlation of circRNA abundance with the
proliferation of liver regeneration (LR) has recently been clarified and the

This article is protected by copyright. All rights reserved


comprehensive circRNA profiles served as candidates for nonalcoholic


steatohepatitis (NASH) diagnosis have been discussed. Recently, many reports
supported circRNAs had special effects on the occurrence and development of liver
disease (Hou and Zhang, 2017). Thereby, this study will focus on the foundation of
revealing the functional roles of circRNAs during the pathological process of liver
diseases, especially in HCC. Remarkably, accumulating datas showed a large
number of circRNAs were differentially expressed in HCC tissues, which indicated
these circRNAs might be singularly regulated and exert a potential function in the
process of HCC (Yao et al., 2017a). The expression of circRNAs in HCC were
listed in Table 1.

The oncogenic or tumor suppressive roles of circRNA

Cdr1as (cerebellar degeneration-related protein 1 antisense RNA), also named as


ciRS-7, is one of the first known circRNAs. The Cdr1as gene represents
naturally-occurring RNAs, which are transcribed in antisense to CDR1 (cerebellar
degeneration-related protein 1). They are naturally present in cytoplasm and primarily
produced in brain regions (Memczak et al., 2013). Hitherto, Cdr1as is one of the best
known circRNAs, with comprehensive abilities to regulate various diseases, such as
cancer, diabetes, atherosclerosis and neurodegenerative diseases. Recently, Yu L et al.
focused on the regulatory role of Cdr1as in HCC, surprised to find the expression
level of Cdr1as was up-regulated in (74%, 26/35) HCC tissues compared with its
normal hepatocytes. Whats more, Cdr1as silencing could suppress the HCC tissues
(SMMC-7721 and HepG2) proliferation measured by CCK-8 assay, which obviously
indicated that Cdr1as might act as an oncogene in HCC (Yu et al., 2016). Interestingly,
further study detected an inversely result that the Cdr1as level in more than half of
HCC tissues (65/108, 60.2 %) were down-regulated when compared with their
adjacent non-tumor tissues (Xu et al., 2017). Taken together, the exact function of
Cdr1as (up-regulated or down-regulated in HCC tissues) remains controversial. But it
is certainly confirmed the expression level of Cdr1as is closely related to HCC. More
extensive studies are still required to detect whether Cdr1as could become a
promising biomarker.
CirZKSCAN1 (circBase ID: hsa_circ_0001727), is a covalently linked 668-nt
circular RNA, combined with exons 2 and 3 of ZKSCAN1. ZKSCAN1, is a zinc
finger family gene, also named as ZNF139, playing regulatory roles for numerous
genes at the transcriptional level and closely relevant to tumorigenesis, metastasis,
and drug tolerance (van Dekken et al., 2009). Currently, the role of circZKSCAN1 in
the regulation of HCC cell growth, migration and invasion as well as the potential
mechanism were investigated. In a novel study, differential expression of
cirZKSCAN1 was observed, based on qRT-PCR analyses of 102 HCC tissue samples
and paired adjacent non-tumorous liver tissues. The results showed circZKSCAN1
was significantly down-regulated in HCC cell lines (Huh7, SMMC-7721, BEL-7402,
HepG2 and Hep3B). Additionally, the result of MTS

This article is protected by copyright. All rights reserved


(3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetr
azolium) assay revealed that over-expression of cirZKSCAN1 effectively inhibited
cell proliferation in SMMC-772 cells. Moreover, knockdown of cirZKSCAN1 could
effectively accelerate cell proliferation, migration and invasion. Particularly, the
expression levels of Caspase 3, RAC2, and EFNA3 in SMMC-7721 cells were
affected by over-expression and knockdown treatments of cirZKSCAN1, which are
important factors in the intrinsic apoptosis pathway. Similar expression patterns were
detected in cell proliferation-related genes, including CCND1, TGFB1, CXCR4,
ITGB4 surviving (Yao et al., 2017b). Based on the emerging evidence, cirZKSCAN1
is involved in the progression of HCC, acting as a competitive inhibitor to maintain
the endogenous RNAs and regulating the expression of tumor cell proliferation and
metastasis-related genes. Furthermore, this trend has gradually been observed that
cirZKSCAN1 can be used as a diagnostic biomarker of HCC, which provide a novel
basis for the early diagnosis or treatment for human HCC.
Shang X et al. identified hsa_circ_0005075 (located on
chromosome1:21377358-21415706) in 61 differentially expressed circRNAs, as the
expression of hsa_circ_0005075 present significant associations with some
clinicopathological factors in HCC patients. hsa_circ_0005075 is a vital component of
eIF4F (cap-dependent translation regulation complex), with the gene symbol of
eIF4G3 (initiation factor 4 gamma 3). Studies found the hsa_circ_0005075 expression
level was apparently up-regulated in HCC tissues and corresponding non-tumorous
tissues as well as hsa_circ_0005075 expression was associated with tumor size.
Intriguingly, higher hsa_circ_0005075 expression showed HCC tumors with a larger
size, which indicated hsa_circ_0005075 playing a key role in promoting tumor
growth. Moreover, hsa_circ_0005075 was predicted to participate in the biological
process of cell adhesion by using GO and pathway analysis, which was involved in
cancer cell proliferation and metastasis (Shang et al., 2016; Yao et al., 2017a).
According to the research above, it is reasonable to speculate that a high expression
level of hsa_circ_0005075 in HCC tissues is possibly correlated with tumor
progression and hsa_circ_0005075 may be utilized as a potential biomarker for HCC
with a high degree of specificity, accuracy and sensitivity.
Further study focused on a circRNA, hsa_circ_0001649 (ID: hsa_circ_0001649),
selected from circBase. hsa_circ_0001649 was reported to be associated with HCC in
circ2Traits (Li et al., 2017b). Corresponding report showed hsa_circ_0001649 is
produced at the SHPRH gene locus with exon 26-29. SHPRH is a member of the
SWI/SNF family of ATP ases/helicases, as a putative tumor suppressor gene, mutating
in an amount of cancer cell lines (Unk et al., 2006). Therefore, probably hypothesis
claimed hsa_circ_0001649 may perform as an important deterrent to carcinogenesis
and mutagenesis in human cancers through targeting the SHPRH gene locus. In
addition, the expression level of hsa_circ_0001649 was detected in 89 samples of
HCC and paired adjacent liver tissues, the result showed that hsa_circ_ 0001649
expression was significantly down-regulated in HCC cell lines. Notably, after
knockdown of hsa_circ_0001649 with siRNA in immortalized HCC-LM3 and

This article is protected by copyright. All rights reserved


MHCC-97L, the expression levels of several matrix metallopeptidases (MMPs) 9, 10,


2, 13, and 7 were significant increased (Qin et al., 2016). MMPs have been reported
to act as key roles in promoting the metastasis of HCC and served for the
development of novel therapeutic approaches and molecular markers to inhibit HCC
metastasis (Sternlicht and Werb, 2001; Stetler-Stevenson, 1990). Particularly, almost
all MMPs investigated is up-regulated in HCCs. For instance, MMP-2 has been
suggested to direct the tumor-related cells invasion, as it is commonly over-expressed
in tumor masses with high abilities to downgrade BM macromolecules. Bramhall et al.
reported MMP-2 abundantly exist in the tumor stroma of pancreatic adenocarcinoma
and MMP-2 also mainly immuno-localized to tumor cells in HCC (Chen et al., 2016b;
Daniele et al., 2014). Whats more, MMP-9 expressed more frequently in HCC
tumors than in corresponding noncancerous tissues, which may become a promising
inhibitor and make great contributions to the development of HCC metastasis, like
colony formation, cell proliferation, migration and invasion (Chen et al., 2012; Wu et
al., 2015). Consequently, the interaction between hsa_circ_000_1649 and MMPs
gives us more profound understanding of HCC treatment, though the subsistent
studies are quite rare and further researches are still needed. The targets of circRNAs
in HCC cell lines were listed in Table 2.
hsa_circ_0005986 (located on chromosome1: 1405749414068652), is one of
the up-regulated circRNAs and its related gene symbol is PRDM2 (PR/SET Domain
2). Recently, a novel study showed hsa_circ_0005986 was down-regulated in HepG2,
Huh7, SMMC7721, MHCC97H, MHCC97L and HCCLM3 cells. Specially, the
expression level of hsa_circ_0005986 in MHCC97H was significantly lower than that
in MHCC97L. MHCC97H and MHCC97L are derived from their parental cell line
MHCC97, which provides standard models for comparative proteome analysis of
HCC metastasis associated proteins (Ding et al., 2004). Moreover, hsa_circ_0005986
silencing could accelerate cell proliferation in Huh7 and HepG2 cells and flow
cytometry also demonstrated the hsa_circ_0005986 down-regulation could promote
the G0/G1 to G2 phase transition in HCC cells (Fu et al., 2017a). The G0/G1 switch
gene 2 (G0S2) was initially detected in blood mononuclear cells following induced
cell cycle progression and hyper-methylated in several human cancer cell lines as well
as in squamous lung, neck and head cancers. Previous evidence revealed that G0S2
expression was up-regulated in chronic hepatitis. Moreover, experiments on wild type
mice implied that G0S2 might be a significant regulator hepatic gene during HCC
cells proliferation by activating PPAR- (PPAR- is well defined as the key regulator
of fatty acid catabolism in the liver) (Heckmann et al., 2013). Generally, the data
above suggested that down-regulation of hsa_circ_0005986 were involved in HCC
progression, especially in the acceleration of cell proliferation.

The function as microRNA sponges

CircRNAs are typically expressed noncoding RNAs, while their biogenesis and
chief functions are not well-understood. A couple of potential functions of circRNAs

This article is protected by copyright. All rights reserved


have been increasingly discovered during the lengthy biological evolution. Observed
in the structure of circRNAs, they have numerous miRNA binding sites which can be
attached to miRNAs like a sponge (Hansen et al., 2013a; Kulcheski et al., 2016).
Some studies proved that circRNAs could function as miRNA sponges to modulate
alternative transcription or splice as well as enhance their abilities for regulating the
expression of parental genes. Therefore, figuring out the accurate behavior of
circRNAs functioning as miRNA sponges have been a focused point once upon a time
(Cortes-Lopez and Miura, 2016; Fan et al., 2017). Examples, such as circRNA-CER
is the sponge for miR-136 (Liu et al., 2016b), testis-specific circRNA (sex
determining region Y) is the sponge for miR-138 (Granados-Riveron and
Aquino-Jarquin, 2014), and circRNA-ITCH is the sponge for miR-7, miR-17, and
miR-214 (Kulcheski et al., 2016). Notably, Cdr1as is a super sponge for miR-7,
possessing more than 70 conventional binding sites (Hansen et al., 2013b). Overview
of the molecular functions of circRNA was showed in Figure 2. Previous studies
demonstrated that Cdr1as is a powerful miR-7 inhibitor/sponge in developing
midbrain of zebrafish, directly regulate the function of miRNAs and its collaboration
genes (Hansen et al., 2013a). In the study conducted by Yu L et al., the authors
showed the level of miR-7 expression was down-regulated in 66% (23/35) HCC cell
lines (Hep3B, HepG2, Bel7402 and SMMC-7721) compared with the HL-7702 cells.
Furthermore, over-expressed miR-7 could not only suppress the proliferation and
invasion in SMMC-7721 and HepG2 cells, but also inhibit the expression of CCNE1
and PIK3CD (Yu et al., 2016). Both CCNE1 and PIK3CD are novel targets of miR-7
in HCC cells. CCNE1 is an important cell cycle regulator, playing a downstream role
in inducing HCC cells cycle arrest in G1/S transition (Zhang et al., 2014). PIK3CD
was found over-expressed in QGY-7703 with L-02 cells and positively inhibited
tumorigenesis and cancer metastasis (Liang et al., 2015). On the other hand, Cdr1as
expression was up-regulated in HCC tissues and knockdown of Cdr1as dramatically
promoted the expression of miR-7 as well as its target gene CCNE1 and PIK3CD (Yu
et al., 2016). Obviously, the results above suggested that the decreased distribution of
Cdr1as finally devoted to decelerating HCC cell proliferation and invasion with the
significant interaction with miR-7.
Whats more, circRNAs are supposed to be the key regulators in the pathological
process of HCC and strikingly influence the activities of various liver diseases. Thus,
it was not surprised to see the HCC-stage-expressive characteristics of
hsa_circ_0004018 performed from chronic hepatitis to cirrhosis and eventually
leading to HCC. Certainly, the level of hsa_circ_0004018 in hepatitis tissues, cirrhosis
tissues or HCC tissues were differently expressed (Fu et al., 2017b). Besides, it was
reported that the expression level of hsa_circ_0005986 was correlated with chronic
hepatitis B family history as well as microvascular invasion, tumor diameters and
Barcelona Clinic Liver Cancer (BCLC) stage (the BCLC staging system was
proposed as a standard for the assessment of prognosis in Europe) (Fu et al., 2017a).
According to these observations, circRNAs seems to challenge their determined
regulatory roles during the differential stages of liver diseases. On the other hand, the

This article is protected by copyright. All rights reserved


amounting evidences indicated circRNAs might act as competitive ceRNAs to


regulate the function of miRNAs (Zheng et al., 2016). Exemplification confirmed that
miR-150 was closely relevant to nine circRNAs and miR-382 was correlated with 22
circRNAs. In addition, miR-150 regulated cell proliferation by targeting VEGF
(Vascular endothelial growth factor), while miR-382 negatively regulated PTEN
(Phosphatase and tensin homolog) expression and increased Akt (Protein Kinase B)
phosphorylation during the procession of LR (Bei et al., 2016; Liu et al., 2013). The
results above indicated that circRNAs may regulate rat LR by binding up to miRNAs
and its associated gene expression (Li et al., 2017a). In further intensive study,
circRNA- miRNA-mRNA pathways were constructed for investigating the effect of
the crucial regulation pathways in NASH models, following the composition
molecules with circRNA_002581-miR-122-Cpeb1, circRNA_002581-miR-122-Plp2,
circRNA_002581-miR-122-Slc1a5, and circRNA_007585-miR-326-UCP2, which
were progressively involved in certain pathophysiologic processes when individually
analyzed (Jin et al., 2016). For example, miR-122 was a key therapeutic target factor
in liver diseases, while miR-326 was validated as one of the independent prognostic
predictor of HCC (Akuta et al., 2016; Zhang et al., 2015). Concerning mRNAs,
Cpeb1 was related to angiogenesis in chronic liver diseases and involved in induction
of insulin resistance, Plp2 was likely to increase ER-stress (Endoplasmic reticulum
stress) induced neuronal apoptosis while Slc1a5 was combined with hepatic
glutamine uptake (Calderone et al., 2016; Hassanein et al., 2013; Ma et al., 2014).
Additionally, activated UCP2 participated in NASH through mitochondrial proton
leak induction (Xu et al., 2012; Xu et al., 2013). Summarizing all the previous
researches, circRNAs are promisingly as diagnostic or prognostic predictor for NASH
and the circRNA-miRNA-mRNA pathway may provide a new basis for refining the
detailed mechanism for NASH (Jin et al., 2016). CircRNAs function as microRNA
sponges by targeting related genes were showed in Figure 3.
Taken together, studies on the remarkable circRNA-miRNA-mRNA pathways
would not only provide novel clues for the pathogenesis of liver diseases, especially
in HCC patients, but also establish a mature platform for further investigations in
other diseases. CircRNAs function as microRNA sponges in HCC were listed in Table
3.

This article is protected by copyright. All rights reserved


Regulation roles of circRNAs in HCC

CircRNAs were first discovered in mouse testis in 1993 by Capel B and


colleagues (Dou et al., 2016). But the information on the mechanism of circRNAs and
their multiple aspects of precise biological function in HCC cells proliferation,
metastasis or apoptosis remains largely unknown.
There are several signaling pathways involved in pathological mechanism of
cancer, such as the MAPK/ERK1/2, AMPK, Bcl2/beclin1 signaling pathways and
PI3K/AKT/mammalian target of rapamycin (mTOR) (Liu et al., 2016a). The
PI3K/AKT/mTOR signaling pathway plays an important role in the progression of
cancers, including colorectal cancer, small cell lung cancer (SCLC) (Chen et al.,
2016a; Costa et al., 2015; Li et al., 2016b). Moreover, the PI3K/AKT/mTOR
signaling pathway is becoming increasingly difficult to ignore its investigation into
the mechanism of circRNAs. At the same time, recent studies supported the
standpoints that the PI3K/AKT/mTOR pathway voluntarily participated in the
initiation, development and metastasis of HCC by activating S6 kinase, which is one
of the crucial G1/S checkpoints of the cell cycle and tumor-related cells metastasis
and invasion (Janku et al., 2014; Xu et al., 2017; Zhu et al., 2017a). Therefore, the
variety of researches indicated the PI3K/AKT/mTOR signaling pathway was able to
be a promising target with great respect to its frequent dysregulation in HCC as well
as its fundamental roles in regulating cell proliferation, migration and angiogenesis.
Furthermore, deep research in 46 HCC samples showed the differential
expressed Cdr1as intervene in the PIK3CD/p70S6K/mTOR pathway by sequestering
miR-7-targeted genes, PIK3CD (phosphoinositide3-kinase catalytic subunit delta) and
p70S6K (Ribosomal protein S6 kinase beta-1), both of which were identified as
integral components in PIK3CD/p70S6K/mTOR pathway. However, the relationship
between Cdr1as and mTOR (Mechanistic target of rapamycin) in HCC still remains a
mystery. Respectively, the final conclusion still needs strengthened by further vivo
and vitro experiments.

Potential clinical application of circRNAs in HCC-the biomarker role

With the rapid improvement of diagnostic strategies and treatment approaches,


HCC patients can be identified at an early stage, exposing to radical surgeries with a
favorable prognosis. However, many patients with HCC missed the best surgical
opportunities with poor survival rates in the early stages, resulting in negligence of
current diagnostic standards. Thus, there always remains a high mortality rate in HCC
(Omino et al., 2017; Shang et al., 2016). Indeed, the involvement of circRNAs in
HCC progression has recently became a widely-concerned focus among scientific
researchers. Traditionally, alpha-fetoprotein (AFP) was extensively employed as a
high-risk factor for the observation and diagnosis of HCC via serum test, despite the
clinical diagnostic value of AFP is limited (Wan et al., 2014). Officially, the 2010
guidelines of the American Association for the Study of Liver Diseases (AASLD)

This article is protected by copyright. All rights reserved


stopped recommending AFP for screening. Therefore, it is imperative to discover


more competent biomarkers to increase the rate of early diagnosis of HCC for
improving the therapeutic effect and prognosis on HCC patients (Morgul et al., 2016;
Yan et al., 2015).
Present reports showed up-regulated Cdr1as expressed in 52.17 % (24/46) HCC
tissues and the expression of Cdr1as was significantly correlated with younger age
and several other clinicopathological characteristics of HCC patients. Furthermore,
the same tendency was observed in patients whose AFP level 400 ng/ml,
suggesting that Cdr1as is a valuable biomarker for HCC treatment (Xu et al., 2017).
Besides, microvascular invasion (MVI) is a canonical qualification for intra-hepatic
tumor dissemination, usually undetectable by preoperative imaging. In addition, series
clinical studies verified the identification with MVI, which can be utilized as a high
risk factor of cancer recurrence and poor prognosis after resection of HCC (Chou et
al., 2012; Jakhete et al., 2016). Compared with the risk factors, the up-regulated
expression of Cdr1as in HCC tissue was the most independent factors of MVI, with
the receiver-operating characteristic (ROC) curve showing the area under the ROC
curve (AUC) was 0.68 (95 % CI 0.58, 0.79) at the cutoff value of 0.135. According to
the data above, Cdr1as was significantly expressed in the deterioration and metastasis
of HCC with great potential to be a risk factor for predicting MVI, which provided
new approaches for HCC treatment (Xu et al., 2017).
As was discussed above, both hsa_circ_0005075 and hsa_circ_0001649 were
expressed in HCC tissues, with totally opposite expression. Similar to
hsa_circ_0005075, the level of hsa_circ_0001649 expression was correlated with
HCC tumor size and HCC tumors with larger size showed lower level of
hsa_circ_0001649 expression, indicating the hsa_circ_0001649 might play a vital role
in promoting tumor growth. Furthermore, welch t-test examined that tumor embolus
was significantly associated with hsa_circ_0001649 expression, which reflected the
potential correlations between hsa_circ_0001649 expression and metastasis
possibility of cancer cells (Qin et al., 2016). The diagnostic effect of
hsa_circ_0001649 was examined by ROC curve in the distinguished HCC tissues and
the AUC was 0.63. As for hsa_circ_0005075, the AUC was 0.94. In conclusion,
whether hsa_circ_0001649 or hsa_circ_0005075 can be adapted according to the
characteristics of each individual patient (Shang et al., 2016).
Additionally, decreased expression of hsa_circ_0004018 was diversely
associated with tumor differentiation, diameters, TNM (tumor-node-metastasis) stage
and BCLC stage. Respectively, the AUC was 0.848 (95% CI=0.8030.894), the
sensitivity and specificity were 0.716 and 0.815 and the Youden index was 0.531 (Fu
et al., 2017b). All of these clinical therapy data firmly confirmed the possibilities that
hsa_circ_0004018 can be used as a promising biomarker matched up to the current
trends and standards in the treatment of HCC.
CircRNAs were firstly used as potential biomarkers in Drosophila for a long
history (Chen et al., 2015; Yao et al., 2017a). After that, studies found circRNAs can
be easily detected in clinical standard blood samples, acting as molecular biomarkers

This article is protected by copyright. All rights reserved


for disease diagnosis or high efficient miRNA regulators. Therefore, the importance
of circRNA was gradually recognized (Jin et al., 2016; Memczak et al., 2015).
Summarizing the observations above, we could find circRNAs in different profiles
can be promising biomarkers to modulate molecular differences in HCC for the
earliest diagnosis, which may be helpful in the selection of the most appropriate
treatment and gain valuable therapy time for HCC patients.

CircRNAs in liver diseases

The liver is a vital organ and supports almost every other organ in the body,
acting as a major integrator of metabolism. Because of its multidimensional functions,
the liver is essential to many diseases (Minemura and Shimizu, 2015). The estimated
prevalence of liver diseases in the general population in North America alone is
2030% (Fazel et al., 2016; Goh and McCullough, 2016). In China, the incidence and
mortality of liver diseases were rapidly increasing and the number of deaths caused by
liver diseases were presented in the top 2 among all types of cancers in 2014 (Wang et
al., 2014; Zhai et al., 2016). Most cases of liver diseases naturally originate from
hepatitis, which is well known as a dynamic disease, commonly associated with
infection/inflammation or cellular toxicity/injury factors, such as HBV and HCV
(Sanarico et al., 2016). As the inflammation progresses, the conditions get worse and
then probably develop into liver fibrosis. Liver fibrosis is a reversible wound-healing
response to any etiological hepatic injuries and it is supposed that the main causes of
liver fibrosis are alcohol, NASH and chronic hepatitis C infection (Guo et al., 2015;
Lurie et al., 2015). The risk of liver-related mortality exponentially increases during
the fibrosis stage. Furthermore, once the liver fibrosis is out of control, experiencing
variable periods of immune activity versus quiescence will lead to the deterioration of
cirrhosis and HCC in a member of patients (Llovet et al., 2016). However, the
development of HCC is a complicated pathological process relevant to the different
stages of hepatitis, liver fibrosis or hepatic cirrhosis in the lifelong procession of liver
diseases. Therefore, it is indispensable to find new approaches for the treatment of
liver diseases, specially, cancer biomarkers may be one of the most practical methods.
Relevant researches about the relationship between circRNAs and liver diseases
were largely conducted. Jin X et al. established mice models with NASH and
discovered a large number of dys-regulated circRNAs in NASH group, including 69
up and 63 down regulated circRNAs compared with control group (Jin et al., 2016).
Whats more, further experiment was designed to verify the regulatory role of huge
amount of circRNAs in rat LR. LR plays a crucial role in various liver-associated
diseases (Kwon et al., 2015; Yi et al., 2016). Following partial hepatectomy, activated
hepatocytes will enter cell proliferation to restore the original liver mass and then
regenerate back to its primary size, this complex process formally regulated by
growth factors, cytokines and non-coding RNAs. Li L et al. found 159 circRNAs of
2412 corresponding linear transcripts of circRNAs, were critically contributed to the
process of substance metabolism, energy metabolism as well as regulation of

This article is protected by copyright. All rights reserved


hepatocyte proliferation during the early stage of rat LR, which indicated that
circRNAs may function as a scaffold for RNA binding proteins applying its biological
mechanisms like protein transcription (Li et al., 2017a). One of a distinguished
circRNA, Circ-FOXO3, was detected to be interacted with the transcription factor
E2F1, the anti-senescent protein ID-1 as well as the anti-stress proteins HIF1a and
FAK, making these proteins loss their function of anti-senescent and anti-stress by
retaining them in cell cytoplasm (Yang et al., 2016b). Besides, the circRNAs,
ci-ankrd52, was associated with elongation Pol II machinery, acting as a positive
regulator in Pol II transcription (Zhang et al., 2013). Furthermore, the novel study
examined the roles of individual circRNAs. It was surprised to discover the
expression of four circRNAs, circ15, circ1366, circ2077 and circ432 participated in
the critical networks of substance metabolism, energy metabolism, and hepatocyte
proliferation, consisting with their linear transcripts GOT1, TNFRSF21, FN1 and
MAPK14 (Li et al., 2017a; Pan et al., 2012). Therefore, those available researches
fully specified the important function of circRNAs in rat LR, which inferred the great
association between circRNAs and liver diseases. Linear transcript of circRNAs in
liver diseases were listed in Table 4.
In sum, additional studies are also required, not only in rat, but in human liver
cells to further clarify the relationship between circRNAs and liver diseases to address
this interesting hypothesis that circRNAs play a crucial role in liver diseases
formation.

Future expectations

Besides, the relationship between miRNA and circRNA deserve to be a


significant part of epigenetics. Additional experiments should be conducted to test the
two distributions of circRNA acted on other cancer types.
Currently, there is a growing interest in developing nanocarriers to deliver
siRNAs or miRNAs, carrying chemotherapeutic drugs to make synergistic effects by
targeting different therapeutic mechanisms (Deng et al., 2014). The nanocarrier-based
miRNA anti-cancer therapies rapid developed with the aim of improving clinical and
survival outcome, either isolated or in combination with ordinary chemotherapeutic
drugs. Therefore, recent studies verified the occurrence of novel lipid-coated cisplatin
nanoparticles co-loaded with microRNA-375 (NPC/miR-375) as a promising
approach for chemotherapy insensitive HCC treatment, as the NPC/miR375
nanoparticles were expected to decelerate cell proliferation and potentiate the
anti-tumor effect of cisplatin in antagonistic HCC cells (Guo et al., 2013; Ho et al.,
2012). It may be reasonable to explain the co-delivery system can be generalized to a
broad spectrum of circRNAs. As it mentioned above, circRNAs act as a novel target
for HCC treatment. Due to the specialty of clinical potential, low toxicity and the ease
of production, co-delivery nano circRNAs represent a valuable approach to reverse
drug resistance of cancer cells, enhancing the efficacy of chemotherapeutic drugs to
improve overall treatment outcomes (Yang et al., 2016a). This filed is worthy of

This article is protected by copyright. All rights reserved


widely attention.
Reference to recent study, exosomes are 40100nm extracellular vesicles (EV),
enriched in endosome-derived components which are secreted in pathological and
physiological conditions by different type of cells (Mittelbrunn and Sanchez-Madrid,
2012). Exosomes play an important role in intercellular communication, especially in
their relevant liver cells. In the liver, exosomes are normally secreted by hepatocytes,
immune cells (i.e., T and B cells, Kupffer cells), nonparenchymal liver cells (i.e.,
stellate cells) and the cellular interplay modulated by these vesicles, holding
important functions in liver homeostasis (Zhang and Wang, 2015). When exosomes
secreted by primary hepatocytes, they would activate the stellate cells by means of
specific RNA cargoes. If exosomes secreted by hepatic stellate cells, they would be
involved in the progress of liver fibrosis (Qu et al., 2015b). Whats more, EVs
released from hepatocytes leading to a macrophages inflammatory phenotype.
Obviously, the above results firmly confirmed the characterization of exosomes in
HCC cells (Clevers, 2011; Santangelo et al., 2017). With more deepening research of
exosomes, miRNAs are enriched in exosomes released from HCC cells, Valadi H et al.
discovered a novel mechanism of exosome-mediated in transfer of microRNAs and
mRNAs. Liu W et al. identified four miRNAs (miRNA-200a, miRNA-122,
miRNA-21 and miRNA-10b) were closely associated with the expression of
exosomes, which combined with circulating miRNAs and exosomes, serving as
promising biomarkers for cirrhosis discrimination and HCC diagnosis (Liu et al.,
2015). Comparing to circRNAs, Dou Y et al. detected circRNAs were existed in the
exosomes of human colon cancer cell lines, HCT116 and HKe3 (Dou et al., 2016).
Moreover, circRNAs were also found in secreted extracellular-vesicles, sometimes,
circRNAs were highly abundant in exosomes than cells. For example, FISH analysis
showed circZKSCAN1 was most likely located in the cytoplasm and greatly enriched
in EVs preparations of HeLa cells, which revealed the stable cirZKSCAN1 was able
to convey messages among other cells via vesicles (Colombo et al., 2013).
Furthermore, Lasda and Parker et al. found circRNAs are enriched in EV preparations
over their linear counterparts (conventionally spliced RNAs originating from the same
gene) compared to the producing cells and circRNAs can be eliminated from cells
into extracellular space by EVs (Lasda and Parker, 2016). Especially, CDR1as was
also detected in EV preparations (Hansen et al., 2013a). An additional possibility
shows that the functional circRNAs in some cases may be packaged into EVs with the
purpose of cell to cell communication. Thus, it is nevertheless to speculate that
circRNAs like Cdr1as adjust the specific functions to their sponged miRNA status
from the source cells to other cells in EVs and modulate the progress of HCC.

This article is protected by copyright. All rights reserved


Conclusion

From HCC statistics in China and other parts of the world, HCC remains a major
threat to human health. The likelihood of developing HCC is increasing with a
prevalent tendency. The severe fact allowed researchers to pay attention on HCC and
more researches will be made. Among these researches, a large number of evidences
pointed the comprehensive profile of circRNAs expression was associated with HCC
and circRNAs might have special effects on HCC progression. For example,
upregulated circRNA can act as a risk factor of MVI in HCC (Xu et al., 2017).
Additionally, Yu L et al. found CCNE1 and PIK3CD in oncogenesis might act as
tumor suppressor genes, demonstrating CCNE1 and PIK3CD might play a key role in
the pathogenesis of HCC, circRNAs perhaps involved in the regulatory mechanism of
HCC (Yu et al., 2016). After Qin et al found that hsa_circ_0001649 was
downregulated in HCC tissues, more and more researchers focused on circRNAs and
HCC. Much more detailed evidences fully proved the potential of circRNAs being
used as a HCC biomarker. For instance, the above experiments showed different
distribution of circRNAs played different roles in cancer progression and miR-7 was
reported to dysregulate circRNAs, would circRNAs be regulated by several other
miRNAs? When crosstalk else influence factors, does the expression of circRNAs
change in the pathogenesis of HCC? It is also not clear whether circRNAs play
critical roles in HCC or manipulation of circRNAs have beneficial effects on the
intervention with HCC. By advanced techniques in further studies, such as
next-generation sequencing and microarray, the mysteries of circRNAs regulation in
cancer and other diseases will be revealed. The most appropriate method of
applications for circRNAs in clinical treatment will eventually be summarized. In the
future, we will continue to focus on this field.

Conflict of interest statement

There is no conflict of interest.

This article is protected by copyright. All rights reserved


Acknowledgments
This project was supported by the National Natural Science Foundation of China (Nos.
81273526, 81473268,81470003), the Provincial Natural Science Research Project of
Colleges and Universities of Anhui Province (No. KJ2016A348), the fund of Anhui
medical university doctoral start research (No. 0601067101), Anhui Medical
University early contact research of clinical medicine(2015-ZQKY-47), Guangdong
Province Science and Technology Project (2013B021800164), Sichuan Medical Law
Research Center(YF16-Y22) and Humanities and Social Science Research Project of
Colleges and Universities of Anhui Province (No. SK2016A0482).

This article is protected by copyright. All rights reserved


References

Akuta N, Kawamura Y, Suzuki F, Saitoh S, Arase Y, Fujiyama S, Sezaki H, Hosaka T, Kobayashi M, Suzuki Y,
Ikeda K, Kumada H. 2016. Analysis of association between circulating miR-122 and
histopathological features of nonalcoholic fatty liver disease in patients free of hepatocellular
carcinoma. BMC Gastroenterol 16(1):141.
Araldi E, Suarez Y. 2016. MicroRNAs as regulators of endothelial cell functions in cardiometabolic
diseases. Biochim Biophys Acta 1861(12 Pt B):2094-2103.
Bei Y, Song Y, Wang F, Dimitrova-Shumkovska J, Xiang Y, Zhao Y, Liu J, Xiao J, Yang C. 2016. miR-382
targeting PTEN-Akt axis promotes liver regeneration. Oncotarget 7(2):1584-1597.
Bonizzato A, Gaffo E, Te Kronnie G, Bortoluzzi S. 2016. CircRNAs in hematopoiesis and hematological
malignancies. Blood Cancer J 6(10):e483.
Caiment F, Gaj S, Claessen S, Kleinjans J. 2015. High-throughput data integration of
RNA-miRNA-circRNA reveals novel insights into mechanisms of benzo[a]pyrene-induced
carcinogenicity. Nucleic Acids Res 43(5):2525-2534.
Calderone V, Gallego J, Fernandez-Miranda G, Garcia-Pras E, Maillo C, Berzigotti A, Mejias M, Bava FA,
Angulo-Urarte A, Graupera M, Navarro P, Bosch J, Fernandez M, Mendez R. 2016. Sequential
Functions of CPEB1 and CPEB4 Regulate Pathologic Expression of Vascular Endothelial
Growth Factor and Angiogenesis in Chronic Liver Disease. Gastroenterology 150(4):982-997
e930.
Chen J, Li Y, Zheng Q, Bao C, He J, Chen B, Lyu D, Zheng B, Xu Y, Long Z, Zhou Y, Zhu H, Wang Y, He X, Shi
Y, Huang S. 2017a. Circular RNA profile identifies circPVT1 as a proliferative factor and
prognostic marker in gastric cancer. Cancer Lett 388:208-219.
Chen L, Huang C, Wang X, Shan G. 2015. Circular RNAs in Eukaryotic Cells. Curr Genomics
16(5):312-318.
Chen QY, Jiao DM, Wu YQ, Chen J, Wang J, Tang XL, Mou H, Hu HZ, Song J, Yan J, Wu LJ, Wang Z. 2016a.
MiR-206 inhibits HGF-induced epithelial-mesenchymal transition and angiogenesis in
non-small cell lung cancer via c-Met /PI3k/Akt/mTOR pathway. Oncotarget
7(14):18247-18261.
Chen R, Cui J, Xu C, Xue T, Guo K, Gao D, Liu Y, Ye S, Ren Z. 2012. The significance of MMP-9 over
MMP-2 in HCC invasiveness and recurrence of hepatocellular carcinoma after curative
resection. Ann Surg Oncol 19 Suppl 3:S375-384.
Chen S, Li T, Zhao Q, Xiao B, Guo J. 2017b. Using circular RNA hsa_circ_0000190 as a new biomarker in
the diagnosis of gastric cancer. Clin Chim Acta 466:167-171.
Chen Y, Lin C, Liu Y, Jiang Y. 2016b. HMGB1 promotes HCC progression partly by downregulating p21
via ERK/c-Myc pathway and upregulating MMP-2. Tumour Biol 37(4):4399-4408.
Chou CT, Chen RC, Lee CW, Ko CJ, Wu HK, Chen YL. 2012. Prediction of microvascular invasion of
hepatocellular carcinoma by pre-operative CT imaging. Br J Radiol 85(1014):778-783.
Chung KY, Kemeny N. 2005. Regional and systemic chemotherapy for primary hepatobiliary cancers
and for colorectal cancer metastatic to the liver. Semin Radiat Oncol 15(4):284-298.
Clevers H. 2011. The cancer stem cell: premises, promises and challenges. Nat Med 17(3):313-319.
Colombo M, Moita C, van Niel G, Kowal J, Vigneron J, Benaroch P, Manel N, Moita LF, Thery C, Raposo
G. 2013. Analysis of ESCRT functions in exosome biogenesis, composition and secretion

This article is protected by copyright. All rights reserved


highlights the heterogeneity of extracellular vesicles. J Cell Sci 126(Pt 24):5553-5565.


Cortes-Lopez M, Miura P. 2016. Emerging Functions of Circular RNAs. Yale J Biol Med 89(4):527-537.
Costa C, Pereira S, Lima L, Peixoto A, Fernandes E, Neves D, Neves M, Gaiteiro C, Tavares A, Gil da
Costa RM, Cruz R, Amaro T, Oliveira PA, Ferreira JA, Santos LL. 2015. Abnormal Protein
Glycosylation and Activated PI3K/Akt/mTOR Pathway: Role in Bladder Cancer Prognosis and
Targeted Therapeutics. PLoS One 10(11):e0141253.
Daniele A, Divella R, Quaranta M, Mattioli V, Casamassima P, Paradiso A, Garrisi VM, Gadaleta CD,
Gadaleta-Caldarola G, Savino E, Maci R, Bellizzi A, Fazio V. 2014. Clinical and prognostic role
of circulating MMP-2 and its inhibitor TIMP-2 in HCC patients prior to and after trans-hepatic
arterial chemo-embolization. Clin Biochem 47(3):184-190.
Deng X, Cao M, Zhang J, Hu K, Yin Z, Zhou Z, Xiao X, Yang Y, Sheng W, Wu Y, Zeng Y. 2014. Hyaluronic
acid-chitosan nanoparticles for co-delivery of MiR-34a and doxorubicin in therapy against
triple negative breast cancer. Biomaterials 35(14):4333-4344.
Ding SJ, Li Y, Shao XX, Zhou H, Zeng R, Tang ZY, Xia QC. 2004. Proteome analysis of hepatocellular
carcinoma cell strains, MHCC97-H and MHCC97-L, with different metastasis potentials.
Proteomics 4(4):982-994.
Dong Y, He D, Peng Z, Peng W, Shi W, Wang J, Li B, Zhang C, Duan C. 2017. Circular RNAs in cancer: an
emerging key player. J Hematol Oncol 10(1):2.
Dou Y, Cha DJ, Franklin JL, Higginbotham JN, Jeppesen DK, Weaver AM, Prasad N, Levy S, Coffey RJ,
Patton JG, Zhang B. 2016. Circular RNAs are down-regulated in KRAS mutant colon cancer
cells and can be transferred to exosomes. Sci Rep 6:37982.
Ebbesen KK, Hansen TB, Kjems J. 2016. Insights into circular RNA biology. RNA Biol:1-11.
Fan X, Weng X, Zhao Y, Chen W, Gan T, Xu D. 2017. Circular RNAs in Cardiovascular Disease: An
Overview. Biomed Res Int 2017:5135781.
Fazel Y, Koenig AB, Sayiner M, Goodman ZD, Younossi ZM. 2016. Epidemiology and natural history of
non-alcoholic fatty liver disease. Metabolism 65(8):1017-1025.
Fischer JW, Leung AK. 2017. CircRNAs: a regulator of cellular stress. Crit Rev Biochem Mol Biol
52(2):220-233.
Fu L, Chen Q, Yao T, Li T, Ying S, Hu Y, Guo J. 2017a. Hsa_circ_0005986 inhibits carcinogenesis by acting
as a miR-129-5p sponge and is used as a novel biomarker for hepatocellular carcinoma.
Oncotarget.
Fu L, Yao T, Chen Q, Mo X, Hu Y, Guo J. 2017b. Screening differential circular RNA expression profiles
reveals hsa_circ_0004018 is associated with hepatocellular carcinoma. Oncotarget.
Goh GB, McCullough AJ. 2016. Natural History of Nonalcoholic Fatty Liver Disease. Dig Dis Sci
61(5):1226-1233.
Granados-Riveron JT, Aquino-Jarquin G. 2014. Does the linear Sry transcript function as a ceRNA for
miR-138? The sense of antisense. F1000Res 3:90.
Gruner H, Cortes-Lopez M, Cooper DA, Bauer M, Miura P. 2016. CircRNA accumulation in the aging
mouse brain. Sci Rep 6:38907.
Guo PF, Jin J, Sun X. 2015. Influence of IL10 gene polymorphisms on the severity of liver fibrosis and
susceptibility to liver cirrhosis in HBV/HCV-infected patients. Infect Genet Evol 30:89-95.
Guo S, Wang Y, Miao L, Xu Z, Lin CM, Zhang Y, Huang L. 2013. Lipid-coated Cisplatin nanoparticles
induce neighboring effect and exhibit enhanced anticancer efficacy. ACS Nano

This article is protected by copyright. All rights reserved


7(11):9896-9904.
Guttman M, Rinn JL. 2012. Modular regulatory principles of large non-coding RNAs. Nature
482(7385):339-346.
Hanan M, Soreq H, Kadener S. 2016. CircRNAs in the brain. RNA Biol:1-7.
Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, Damgaard CK, Kjems J. 2013a. Natural RNA
circles function as efficient microRNA sponges. Nature 495(7441):384-388.
Hansen TB, Kjems J, Damgaard CK. 2013b. Circular RNA and miR-7 in cancer. Cancer Res
73(18):5609-5612.
Hassanein M, Hoeksema MD, Shiota M, Qian J, Harris BK, Chen H, Clark JE, Alborn WE, Eisenberg R,
Massion PP. 2013. SLC1A5 mediates glutamine transport required for lung cancer cell growth
and survival. Clin Cancer Res 19(3):560-570.
He J, Xie Q, Xu H, Li J, Li Y. 2017. Circular RNAs and cancer. Cancer Lett 396:138-144.
Heckmann BL, Zhang X, Xie X, Liu J. 2013. The G0/G1 switch gene 2 (G0S2): regulating metabolism and
beyond. Biochim Biophys Acta 1831(2):276-281.
Herceg Z, Paliwal A. 2011. Epigenetic mechanisms in hepatocellular carcinoma: how environmental
factors influence the epigenome. Mutat Res 727(3):55-61.
Ho C, Wang C, Mattu S, Destefanis G, Ladu S, Delogu S, Armbruster J, Fan L, Lee SA, Jiang L,
Dombrowski F, Evert M, Chen X, Calvisi DF. 2012. AKT (v-akt murine thymoma viral oncogene
homolog 1) and N-Ras (neuroblastoma ras viral oncogene homolog) coactivation in the
mouse liver promotes rapid carcinogenesis by way of mTOR (mammalian target of rapamycin
complex 1), FOXM1 (forkhead box M1)/SKP2, and c-Myc pathways. Hepatology
55(3):833-845.
Holdt LM, Stahringer A, Sass K, Pichler G, Kulak NA, Wilfert W, Kohlmaier A, Herbst A, Northoff BH,
Nicolaou A, Gabel G, Beutner F, Scholz M, Thiery J, Musunuru K, Krohn K, Mann M, Teupser D.
2016. Circular non-coding RNA ANRIL modulates ribosomal RNA maturation and
atherosclerosis in humans. Nat Commun 7:12429.
Hou LD, Zhang J. 2017. Circular RNAs: An emerging type of RNA in cancer. Int J Immunopathol
Pharmacol 30(1):1-6.
Hsu MT, Coca-Prados M. 1979. Electron microscopic evidence for the circular form of RNA in the
cytoplasm of eukaryotic cells. Nature 280(5720):339-340.
Hwang YH, Choi JY, Kim S, Chung ES, Kim T, Koh SS, Lee B, Bae SH, Kim J, Park YM. 2004.
Over-expression of c-raf-1 proto-oncogene in liver cirrhosis and hepatocellular carcinoma.
Hepatol Res 29(2):113-121.
Ivanov A, Memczak S, Wyler E, Torti F, Porath HT, Orejuela MR, Piechotta M, Levanon EY, Landthaler M,
Dieterich C, Rajewsky N. 2015. Analysis of intron sequences reveals hallmarks of circular RNA
biogenesis in animals. Cell Rep 10(2):170-177.
Jakhete N, Saberi B, Jonassaint NL, Cosar AM, Luu H, Kim A, Anders RA, Philosophe B, Cameron AM,
Gurakar A. 2016. Microvascular Invasion in Hepatocellular Carcinoma and Liver Transplant.
Exp Clin Transplant 14(Suppl 3):14-18.
Janku F, Kaseb AO, Tsimberidou AM, Wolff RA, Kurzrock R. 2014. Identification of novel therapeutic
targets in the PI3K/AKT/mTOR pathway in hepatocellular carcinoma using targeted next
generation sequencing. Oncotarget 5(10):3012-3022.
Jeck WR, Sorrentino JA, Wang K, Slevin MK, Burd CE, Liu J, Marzluff WF, Sharpless NE. 2013. Circular

This article is protected by copyright. All rights reserved


RNAs are abundant, conserved, and associated with ALU repeats. RNA 19(2):141-157.
Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. 2011. Global cancer statistics. CA Cancer J
Clin 61(2):69-90.
Jin X, Feng CY, Xiang Z, Chen YP, Li YM. 2016. CircRNA expression pattern and circRNA-miRNA-mRNA
network in the pathogenesis of nonalcoholic steatohepatitis. Oncotarget 7(41):66455-66467.
Kulcheski FR, Christoff AP, Margis R. 2016. Circular RNAs are miRNA sponges and can be used as a new
class of biomarker. J Biotechnol 238:42-51.
Kuo YH, Wu IP, Wang JH, Hung CH, Rau KM, Chen CH, Kee KM, Hu TH, Lu SN. 2017. The outcome of
sorafenib monotherapy on hepatocellular carcinoma with portal vein tumor thrombosis.
Invest New Drugs.
Kwon YJ, Lee KG, Choi D. 2015. Clinical implications of advances in liver regeneration. Clin Mol Hepatol
21(1):7-13.
Lasda E, Parker R. 2016. Circular RNAs Co-Precipitate with Extracellular Vesicles: A Possible Mechanism
for circRNA Clearance. PLoS One 11(2):e0148407.
Levine AJ, Oren M. 2009. The first 30 years of p53: growing ever more complex. Nat Rev Cancer
9(10):749-758.
Levrero M. 2006. Viral hepatitis and liver cancer: the case of hepatitis C. Oncogene 25(27):3834-3847.
Li H, Hao X, Wang H, Liu Z, He Y, Pu M, Zhang H, Yu H, Duan J, Qu S. 2016a. Circular RNA Expression
Profile of Pancreatic Ductal Adenocarcinoma Revealed by Microarray. Cell Physiol Biochem
40(6):1334-1344.
Li H, Hu J, Wu S, Wang L, Cao X, Zhang X, Dai B, Cao M, Shao R, Zhang R, Majidi M, Ji L, Heymach JV,
Wang M, Pan S, Minna J, Mehran RJ, Swisher SG, Roth JA, Fang B. 2016b. Auranofin-mediated
inhibition of PI3K/AKT/mTOR axis and anticancer activity in non-small cell lung cancer cells.
Oncotarget 7(3):3548-3558.
Li L, Guo J, Chen Y, Chang C, Xu C. 2017a. Comprehensive CircRNA expression profile and selection of
key CircRNAs during priming phase of rat liver regeneration. BMC Genomics 18(1):80.
Li WH, Song YC, Zhang H, Zhou ZJ, Xie X, Zeng QN, Guo K, Wang T, Xia P, Chang DM. 2017b. Decreased
Expression of Hsa_circ_00001649 in Gastric Cancer and Its Clinical Significance. Dis Markers
2017:4587698.
Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, Zhong G, Yu B, Hu W, Dai L, Zhu P, Chang Z, Wu Q, Zhao Y,
Jia Y, Xu P, Liu H, Shan G. 2015. Exon-intron circular RNAs regulate transcription in the nucleus.
Nat Struct Mol Biol 22(3):256-264.
Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, Zhong G, Yu B, Hu W, Dai L, Zhu P, Chang Z, Wu Q, Zhao Y,
Jia Y, Xu P, Liu H, Shan G. 2017c. Corrigendum: Exon-intron circular RNAs regulate
transcription in the nucleus. Nat Struct Mol Biol 24(2):194.
Liang M, Liu J, Ji H, Chen M, Zhao Y, Li S, Zhang X, Li J. 2015. A Aconitum coreanum polysaccharide
fraction induces apoptosis of hepatocellular carcinoma (HCC) cells via pituitary tumor
transforming gene 1 (PTTG1)-mediated suppression of the P13K/Akt and activation of p38
MAPK signaling pathway and displays antitumor activity in vivo. Tumour Biol
36(9):7085-7091.
Liu D, Zhang Y, Wei Y, Liu G, Liu Y, Gao Q, Zou L, Zeng W, Zhang N. 2016a. Activation of AKT pathway by
Nrf2/PDGFA feedback loop contributes to HCC progression. Oncotarget 7(40):65389-65402.
Liu Q, Zhang X, Hu X, Dai L, Fu X, Zhang J, Ao Y. 2016b. Circular RNA Related to the Chondrocyte ECM

This article is protected by copyright. All rights reserved


Regulates MMP13 Expression by Functioning as a MiR-136 'Sponge' in Human Cartilage


Degradation. Sci Rep 6:22572.
Liu WH, Ren LN, Wang X, Wang T, Zhang N, Gao Y, Luo H, Navarro-Alvarez N, Tang LJ. 2015.
Combination of exosomes and circulating microRNAs may serve as a promising tumor marker
complementary to alpha-fetoprotein for early-stage hepatocellular carcinoma diagnosis in
rats. J Cancer Res Clin Oncol 141(10):1767-1778.
Liu Y, Zhao L, Li D, Yin Y, Zhang CY, Li J, Zhang Y. 2013. Microvesicle-delivery miR-150 promotes
tumorigenesis by up-regulating VEGF, and the neutralization of miR-150 attenuate tumor
development. Protein Cell 4(12):932-941.
Llovet JM, Zucman-Rossi J, Pikarsky E, Sangro B, Schwartz M, Sherman M, Gores G. 2016.
Hepatocellular carcinoma. Nat Rev Dis Primers 2:16018.
Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, Abraham J, Adair T, Aggarwal R, Ahn SY,
Alvarado M, Anderson HR, Anderson LM, Andrews KG, Atkinson C, Baddour LM, Barker-Collo
S, Bartels DH, Bell ML, Benjamin EJ, Bennett D, Bhalla K, Bikbov B, Bin Abdulhak A, Birbeck G,
Blyth F, Bolliger I, Boufous S, Bucello C, Burch M, Burney P, Carapetis J, Chen H, Chou D,
Chugh SS, Coffeng LE, Colan SD, Colquhoun S, Colson KE, Condon J, Connor MD, Cooper LT,
Corriere M, Cortinovis M, de Vaccaro KC, Couser W, Cowie BC, Criqui MH, Cross M,
Dabhadkar KC, Dahodwala N, De Leo D, Degenhardt L, Delossantos A, Denenberg J, Des
Jarlais DC, Dharmaratne SD, Dorsey ER, Driscoll T, Duber H, Ebel B, Erwin PJ, Espindola P,
Ezzati M, Feigin V, Flaxman AD, Forouzanfar MH, Fowkes FG, Franklin R, Fransen M, Freeman
MK, Gabriel SE, Gakidou E, Gaspari F, Gillum RF, Gonzalez-Medina D, Halasa YA, Haring D,
Harrison JE, Havmoeller R, Hay RJ, Hoen B, Hotez PJ, Hoy D, Jacobsen KH, James SL, Jasrasaria
R, Jayaraman S, Johns N, Karthikeyan G, Kassebaum N, Keren A, Khoo JP, Knowlton LM,
Kobusingye O, Koranteng A, Krishnamurthi R, Lipnick M, Lipshultz SE, Ohno SL, Mabweijano J,
MacIntyre MF, Mallinger L, March L, Marks GB, Marks R, Matsumori A, Matzopoulos R,
Mayosi BM, McAnulty JH, McDermott MM, McGrath J, Mensah GA, Merriman TR, Michaud C,
Miller M, Miller TR, Mock C, Mocumbi AO, Mokdad AA, Moran A, Mulholland K, Nair MN,
Naldi L, Narayan KM, Nasseri K, Norman P, O'Donnell M, Omer SB, Ortblad K, Osborne R,
Ozgediz D, Pahari B, Pandian JD, Rivero AP, Padilla RP, Perez-Ruiz F, Perico N, Phillips D, Pierce
K, Pope CA, 3rd, Porrini E, Pourmalek F, Raju M, Ranganathan D, Rehm JT, Rein DB, Remuzzi G,
Rivara FP, Roberts T, De Leon FR, Rosenfeld LC, Rushton L, Sacco RL, Salomon JA, Sampson U,
Sanman E, Schwebel DC, Segui-Gomez M, Shepard DS, Singh D, Singleton J, Sliwa K, Smith E,
Steer A, Taylor JA, Thomas B, Tleyjeh IM, Towbin JA, Truelsen T, Undurraga EA,
Venketasubramanian N, Vijayakumar L, Vos T, Wagner GR, Wang M, Wang W, Watt K,
Weinstock MA, Weintraub R, Wilkinson JD, Woolf AD, Wulf S, Yeh PH, Yip P, Zabetian A, Zheng
ZJ, Lopez AD, Murray CJ, AlMazroa MA, Memish ZA. 2012. Global and regional mortality from
235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global
Burden of Disease Study 2010. Lancet 380(9859):2095-2128.
Lukiw WJ. 2013. Circular RNA (circRNA) in Alzheimer's disease (AD). Front Genet 4:307.
Lurie Y, Webb M, Cytter-Kuint R, Shteingart S, Lederkremer GZ. 2015. Non-invasive diagnosis of liver
fibrosis and cirrhosis. World J Gastroenterol 21(41):11567-11583.
Ma XZ, Bartczak A, Zhang J, He W, Shalev I, Smil D, Chen L, Phillips J, Feld JJ, Selzner N, Levy G,
McGilvray I. 2014. Protein interferon-stimulated gene 15 conjugation delays but does not

This article is protected by copyright. All rights reserved


overcome coronavirus proliferation in a model of fulminant hepatitis. J Virol


88(11):6195-6204.
Manning KS, Cooper TA. 2017. The roles of RNA processing in translating genotype to phenotype. Nat
Rev Mol Cell Biol 18(2):102-114.
Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, Maier L, Mackowiak SD, Gregersen LH,
Munschauer M, Loewer A, Ziebold U, Landthaler M, Kocks C, le Noble F, Rajewsky N. 2013.
Circular RNAs are a large class of animal RNAs with regulatory potency. Nature
495(7441):333-338.
Memczak S, Papavasileiou P, Peters O, Rajewsky N. 2015. Identification and Characterization of
Circular RNAs As a New Class of Putative Biomarkers in Human Blood. PLoS One
10(10):e0141214.
Minemura M, Shimizu Y. 2015. Gut microbiota and liver diseases. World J Gastroenterol
21(6):1691-1702.
Mittelbrunn M, Sanchez-Madrid F. 2012. Intercellular communication: diverse structures for exchange
of genetic information. Nat Rev Mol Cell Biol 13(5):328-335.
Morgul MH, Klunk S, Anastasiadou Z, Gauger U, Dietel C, Reutzel-Selke A, Felgendref P, Hau HM,
Tautenhahn HM, Schmuck RB, Raschzok N, Sauer IM, Bartels M. 2016. Diagnosis of HCC for
patients with cirrhosis using miRNA profiles of the tumor-surrounding tissue - A statistical
model based on stepwise penalized logistic regression. Exp Mol Pathol 101(2):165-171.
Mou T, Zhu D, Wei X, Li T, Zheng D, Pu J, Guo Z, Wu Z. 2017. Identification and interaction analysis of
key genes and microRNAs in hepatocellular carcinoma by bioinformatics analysis. World J
Surg Oncol 15(1):63.
Omino R, Mittal S, Kramer JR, Chayanupatkul M, Richardson P, Kanwal F. 2017. The Validity of HCC
Diagnosis Codes in Chronic Hepatitis B Patients in the Veterans Health Administration. Dig Dis
Sci 62(5):1180-1185.
Pan C, Chen H, Wang L, Yang S, Fu H, Zheng Y, Miao M, Jiao B. 2012. Down-regulation of MiR-127
facilitates hepatocyte proliferation during rat liver regeneration. PLoS One 7(6):e39151.
Peng N, Shi L, Zhang Q, Hu Y, Wang N, Ye H. 2017. Microarray profiling of circular RNAs in human
papillary thyroid carcinoma. PLoS One 12(3):e0170287.
Poon RT. 2009. Differentiating early and late recurrences after resection of HCC in cirrhotic patients:
implications on surveillance, prevention, and treatment strategies. Ann Surg Oncol
16(4):792-794.
Qin M, Liu G, Huo X, Tao X, Sun X, Ge Z, Yang J, Fan J, Liu L, Qin W. 2016. Hsa_circ_0001649: A circular
RNA and potential novel biomarker for hepatocellular carcinoma. Cancer Biomark
16(1):161-169.
Qu S, Yang X, Li X, Wang J, Gao Y, Shang R, Sun W, Dou K, Li H. 2015a. Circular RNA: A new star of
noncoding RNAs. Cancer Lett 365(2):141-148.
Qu Z, Jiang C, Wu J, Ding Y. 2015b. Exosomes as potent regulators of HCC malignancy and potential
bio-tools in clinical application. Int J Clin Exp Med 8(10):17088-17095.
Rong D, Tang W, Li Z, Zhou J, Shi J, Wang H, Cao H. 2017. Novel insights into circular RNAs in clinical
application of carcinomas. Onco Targets Ther 10:2183-2188.
Rybak-Wolf A, Stottmeister C, Glazar P, Jens M, Pino N, Giusti S, Hanan M, Behm M, Bartok O,
Ashwal-Fluss R, Herzog M, Schreyer L, Papavasileiou P, Ivanov A, Ohman M, Refojo D, Kadener

This article is protected by copyright. All rights reserved


S, Rajewsky N. 2015. Circular RNAs in the Mammalian Brain Are Highly Abundant, Conserved,
and Dynamically Expressed. Mol Cell 58(5):870-885.
Sabin LR, Delas MJ, Hannon GJ. 2013. Dogma derailed: the many influences of RNA on the genome.
Mol Cell 49(5):783-794.
Salzman J, Chen RE, Olsen MN, Wang PL, Brown PO. 2013. Cell-type specific features of circular RNA
expression. PLoS Genet 9(9):e1003777.
Sanarico N, D'Amato S, Bruni R, Rovetto C, Salvi E, Di Zeo P, Chionne P, Madonna E, Pisani G,
Costantino A, Equestre M, Tosti ME, Cenci A, Maggiorella MT, Sernicola L, Pontali E, Pansera A,
Quattrocchi R, Carbonara S, Signorile F, Surace LA, Federzoni G, Garlassi E, Starnini G,
Monarca R, Babudieri S, Rapicetta M, Pompa MG, Caraglia A, Ensoli B, Ciccaglione AR, Butto S.
2016. Correlates of infection and molecular characterization of blood-borne HIV, HCV, and
HBV infections in HIV-1 infected inmates in Italy: An observational cross-sectional study.
Medicine (Baltimore) 95(44):e5257.
Santangelo L, Battistelli C, Montaldo C, Citarella F, Strippoli R, Cicchini C. 2017. Functional Roles and
Therapeutic Applications of Exosomes in Hepatocellular Carcinoma. Biomed Res Int
2017:2931813.
Shang X, Li G, Liu H, Li T, Liu J, Zhao Q, Wang C. 2016. Comprehensive Circular RNA Profiling Reveals
That hsa_circ_0005075, a New Circular RNA Biomarker, Is Involved in Hepatocellular
Crcinoma Development. Medicine (Baltimore) 95(22):e3811.
Singal AG, El-Serag HB. 2015. Hepatocellular Carcinoma From Epidemiology to Prevention: Translating
Knowledge into Practice. Clin Gastroenterol Hepatol 13(12):2140-2151.
Sternlicht MD, Werb Z. 2001. How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev
Biol 17:463-516.
Stetler-Stevenson WG. 1990. Type IV collagenases in tumor invasion and metastasis. Cancer
Metastasis Rev 9(4):289-303.
Sun L, Liu T, Zhang S, Guo K, Liu Y. 2017. Oct4 induces EMT through LEF1/beta-catenin dependent WNT
signaling pathway in hepatocellular carcinoma. Oncol Lett 13(4):2599-2606.
Takahashi K, Yan IK, Kogure T, Haga H, Patel T. 2014. Extracellular vesicle-mediated transfer of long
non-coding RNA ROR modulates chemosensitivity in human hepatocellular cancer. FEBS Open
Bio 4:458-467.
Tong R, Yang B, Xiao H, Peng C, Hu W, Weng X, Cheng S, Du C, Lv Z, Ding C, Zhou L, Xie H, Wu J, Zheng S.
2017. KCTD11 inhibits growth and metastasis of hepatocellular carcinoma through activating
Hippo signaling. Oncotarget.
Unk I, Hajdu I, Fatyol K, Szakal B, Blastyak A, Bermudez V, Hurwitz J, Prakash L, Prakash S, Haracska L.
2006. Human SHPRH is a ubiquitin ligase for Mms2-Ubc13-dependent polyubiquitylation of
proliferating cell nuclear antigen. Proc Natl Acad Sci U S A 103(48):18107-18112.
van Dekken H, Tilanus HW, Hop WC, Dinjens WN, Wink JC, Vissers KJ, van Marion R. 2009. Array
comparative genomic hybridization, expression array, and protein analysis of critical regions
on chromosome arms 1q, 7q, and 8p in adenocarcinomas of the gastroesophageal junction.
Cancer Genet Cytogenet 189(1):37-42.
Wahid B, Ali A, Rafique S, Idrees M. 2017. New Insights into the Epigenetics of Hepatocellular
Carcinoma. Biomed Res Int 2017:1609575.
Wan HG, Xu H, Gu YM, Wang H, Xu W, Zu MH. 2014. Comparison osteopontin vs AFP for the diagnosis

This article is protected by copyright. All rights reserved


of HCC: a meta-analysis. Clin Res Hepatol Gastroenterol 38(6):706-714.


Wang FS, Fan JG, Zhang Z, Gao B, Wang HY. 2014. The global burden of liver disease: the major impact
of China. Hepatology 60(6):2099-2108.
Wang X, Zhang Y, Huang L, Zhang J, Pan F, Li B, Yan Y, Jia B, Liu H, Li S, Zheng W. 2015. Decreased
expression of hsa_circ_001988 in colorectal cancer and its clinical significances. Int J Clin Exp
Pathol 8(12):16020-16025.
Wu YJ, Neoh CA, Tsao CY, Su JH, Li HH. 2015. Sinulariolide Suppresses Human Hepatocellular
Carcinoma Cell Migration and Invasion by Inhibiting Matrix Metalloproteinase-2/-9 through
MAPKs and PI3K/Akt Signaling Pathways. Int J Mol Sci 16(7):16469-16482.
Xiong XX, Qiu XY, Hu DX, Chen XQ. 2017. Advances in Hypoxia-mediated Mechanisms in Hepatocellular
Carcinoma. Mol Pharmacol.
Xu J, Li N, Wang J, Zhang C, Ding S, Jiao Y. 2012. [Hepatic SIRT1 and UCP2 expressions in rats with type
2 diabetes mellitus and nonalcoholic fatty liver]. Nan Fang Yi Ke Da Xue Xue Bao
32(5):726-729.
Xu J, Li N, Wang J, Zhang C, Ding S, Jiao Y, Zhang J. 2013. [Effect of metformin on the expression of
SIRT1 and UCP2 in rat liver of type 2 diabetes mellitus and nonalcoholic fatty liver]. Zhong
Nan Da Xue Xue Bao Yi Xue Ban 38(9):882-887.
Xu L, Zhang M, Zheng X, Yi P, Lan C, Xu M. 2017. The circular RNA ciRS-7 (Cdr1as) acts as a risk factor of
hepatic microvascular invasion in hepatocellular carcinoma. J Cancer Res Clin Oncol
143(1):17-27.
Xuan L, Qu L, Zhou H, Wang P, Yu H, Wu T, Wang X, Li Q, Tian L, Liu M, Sun Y. 2016. Circular RNA: a
novel biomarker for progressive laryngeal cancer. Am J Transl Res 8(2):932-939.
Yan SR, Liu ZJ, Yu S, Bao YX. 2015. Investigation of the value of miR-21 in the diagnosis of early stage
HCC and its prognosis: a meta-analysis. Genet Mol Res 14(3):11573-11586.
Yang T, Zhao P, Rong Z, Li B, Xue H, You J, He C, Li W, He X, Lee RJ, Ma X, Xiang G. 2016a. Anti-tumor
Efficiency of Lipid-coated Cisplatin Nanoparticles Co-loaded with MicroRNA-375. Theranostics
6(1):142-154.
Yang W, Du WW, Li X, Yee AJ, Yang BB. 2016b. Foxo3 activity promoted by non-coding effects of
circular RNA and Foxo3 pseudogene in the inhibition of tumor growth and angiogenesis.
Oncogene 35(30):3919-3931.
Yao T, Chen Q, Fu L, Guo J. 2017a. Circular RNAs: Biogenesis, properties, roles, and their relationships
with liver diseases. Hepatol Res 47(6):497-504.
Yao Z, Luo J, Hu K, Lin J, Huang H, Wang Q, Zhang P, Xiong Z, He C, Huang Z, Liu B, Yang Y. 2017b.
ZKSCAN1 gene and its related circular RNA (circZKSCAN1) both inhibit hepatocellular
carcinoma cell growth, migration, and invasion but through different signaling pathways. Mol
Oncol 11(4):422-437.
Yi PS, Zhang M, Xu MQ. 2016. Role of microRNA in liver regeneration. Hepatobiliary Pancreat Dis Int
15(2):141-146.
Yu L, Gong X, Sun L, Zhou Q, Lu B, Zhu L. 2016. The Circular RNA Cdr1as Act as an Oncogene in
Hepatocellular Carcinoma through Targeting miR-7 Expression. PLoS One 11(7):e0158347.
Zhai HL, Wang NJ, Han B, Li Q, Chen Y, Zhu CF, Chen YC, Xia FZ, Cang Z, Zhu CX, Lu M, Lu YL. 2016. Low
vitamin D levels and non-alcoholic fatty liver disease, evidence for their independent
association in men in East China: a cross-sectional study (Survey on Prevalence in East China

This article is protected by copyright. All rights reserved


for Metabolic Diseases and Risk Factors (SPECT-China)). Br J Nutr 115(8):1352-1359.


Zhang J, Chong CC, Chen GG, Lai PB. 2015. A Seven-microRNA Expression Signature Predicts Survival in
Hepatocellular Carcinoma. PLoS One 10(6):e0128628.
Zhang X, Hu S, Wang L, Yan B, Zhao J, Yang A, Zhang R. 2014. MicroRNA-7 arrests cell cycle in G1 phase
by directly targeting CCNE1 in human hepatocellular carcinoma cells. Biochem Biophys Res
Commun 443(3):1078-1084.
Zhang Y, Wang XF. 2015. A niche role for cancer exosomes in metastasis. Nat Cell Biol 17(6):709-711.
Zhang Y, Zhang XO, Chen T, Xiang JF, Yin QF, Xing YH, Zhu S, Yang L, Chen LL. 2013. Circular intronic long
noncoding RNAs. Mol Cell 51(6):792-806.
Zhao Y, Alexandrov PN, Jaber V, Lukiw WJ. 2016. Deficiency in the Ubiquitin Conjugating Enzyme
UBE2A in Alzheimer's Disease (AD) is Linked to Deficits in a Natural Circular miRNA-7 Sponge
(circRNA; ciRS-7). Genes (Basel) 7(12).
Zhao ZJ, Shen J. 2015. Circular RNA participates in the carcinogenesis and the malignant behavior of
cancer. RNA Biol:1-8.
Zheng Q, Bao C, Guo W, Li S, Chen J, Chen B, Luo Y, Lyu D, Li Y, Shi G, Liang L, Gu J, He X, Huang S. 2016.
Circular RNA profiling reveals an abundant circHIPK3 that regulates cell growth by sponging
multiple miRNAs. Nat Commun 7:11215.
Zhu H, Liu Q, Tang J, Xie Y, Xu X, Huang R, Zhang Y, Jin K, Sun B. 2017a. Alpha1-ACT Functions as a
Tumour Suppressor in Hepatocellular Carcinoma by Inhibiting the PI3K/AKT/mTOR Signalling
Pathway via Activation of PTEN. Cell Physiol Biochem 41(6):2289-2306.
Zhu M, Xu Y, Chen Y, Yan F. 2017b. Circular BANP, an upregulated circular RNA that modulates cell
proliferation in colorectal cancer. Biomed Pharmacother 88:138-144.
Zimmerman MA, Ghobrial RM, Tong MJ, Hiatt JR, Cameron AM, Hong J, Busuttil RW. 2008. Recurrence
of hepatocellular carcinoma following liver transplantation: a review of preoperative and
postoperative prognostic indicators. Arch Surg 143(2):182-188; discussion 188.

This article is protected by copyright. All rights reserved


Figure legends

Figure 1. CircRNA is formed by backsplicing

Figure 2. Overview of the molecular functions of circRNA. (A) CircRNAs can act as
spongesinteracting with miRNAs. (B) CircRNA can act as the transport of
miRNAs. (C) CircRNAs can enhance the expression of parent genes; ElciRNA can
also enhance the expression of parental genes with U1 snRNP and Pol II. (D)
CircRNA can generate functional proteins.

Figure 3. CircRNAs function as microRNA sponges by targeting related genes.

This article is protected by copyright. All rights reserved


Table 1. The expression of circRNAs in HCC


Expressi Referenc
Sampl Function
circRNA Genome location on es
e in HCC
change (PMID)
HCC
tissue
Biomark 2761445
CDR1as s +
er 3
(74%,
26/35)
Oncogen
e
HCC
circZKSCAN1
tissue Biomake 2821121
(hsa_circ_00017 -
s r 5
27)
(102)
Suppress
or gene
HCC
hsa_circ_00016 chr6:146209155-14621 Biomark 2660039
tissue -
49 6113 er 7
s (89)
Suppress
or gene
HCC
hsa_circ_00050 chr1:21377358-214157 Biomake 2725852
tissue +
75 06 r 1
s (60)
Oncogen
e
HCC
tissue
hsa_circ_00059 chr1:1405749414068 s Biomark 2841021
-
86 652 (80.2 er 1
%,
65/81)
Suppress
or gene
HCC
tissue
hsa_circ_00040 s Biomark 2843066
-
18 (92.2 er 0
%,
94/10

This article is protected by copyright. All rights reserved


2)
Suppress
or gene

This article is protected by copyright. All rights reserved


Table 2. The targets of circRNAs in HCC cell lines


References
circRNA Target Cell lines
(PMID)
CDR1as (ciRS-7) CCNE1, PIK3CD SMMC-7721 27391479
RAC, EFNA3, Caspase 3,
circZKSCAN1 COL3A1, CDH5, MYB, PDK1, SMMC-7721 28211215
BCL2
HCC-LM3 and
hsa_circ_0001649 MMP-9, MMP-10, MMP-13 26600397
MHCC-97L
HCC-LM3 and
hsa_circ_0001649 U2AF65, EIF4A3, UPF1 26600397
MHCC-97L

This article is protected by copyright. All rights reserved


Table 3. CircRNAs function as microRNA sponges in HCC


microRNA References
circRNA cells
sponged (PMID)
HCC cells, SMMC-7221,
CDR1as miR-7 27391479
Hep3B, QGY-7703, HepG2
hsa_circ_0005075 miR-23a HCC cells, HepG2 27258521
hsa_circ_0005075 miR-93 HCC cells, HepG2, SNU449 27258521
hsa_circ_0005075 miR-581 HCC cells, HepG2 27258521
hsa_circ_0001649 miR-182 HCC cells, SK-HEP-1 28185365
HCC cells, HepG2,
hsa_circ_0001649 miR-1297 28185365
SMMC7721

This article is protected by copyright. All rights reserved


Table 4. Linear transcript of circRNAs in liver diseases


liver References
circRNA Gene symbol (Best related linear transcript)
diseases (PMID)
circ013935 Gm21992 (ENSMUST00000178353) NASH 27677588
Carm1 (Coactivator-associated arginine
circ002319 NASH 27677588
methyltransferase 1)
Rpph1 (Ribonuclease P RNA component
circ016983 NASH 27677588
H1)
circ009295 Mpc2 (Mitochondrial pyruvate carrier 2) NASH 27677588
Agap1 ( Ankyrin Repeat And PH Domain
circ017649 NASH 27677588
1)
circ011775 Rn45s (45S pre-ribosomal RNA) NASH 27677588
circ011174 XLOC_010800 (TCONS_00013579) NASH 27677588
circ006982 Ndst3 (N-deacetylase/N-sulfotransferase 3) NASH 27677588
Eefsec (eukaryotic elongation factor,
circ010484 NASH 27677588
selenocysteine-tRNA-specific)
circ011235 Pvt1 (Plasmacytoma variant translocation 1) NASH 27677588
MAPK14 (Mitogen-activated protein kinase Liver
circ432 26747300
14) fibrosis
circ2077 FN1 (Fibronectin 1) HCC 25909171
TNFRSF21 (Tumor necrosis factor receptor
circ1366 LR 28086788
superfamily member 21)
GOT1 (Glutamic-oxaloacetic transaminase
circ15 LR 28086788
1)
circ137 BCL6 (B-cell lymphoma 6 protein) LR 28086788
circ2270 SETD8 (SET domain-containing protein 8) LR 28086788

This article is protected by copyright. All rights reserved


This article is protected by copyright. All rights reserved


Figure 2

This article is protected by copyright. All rights reserved


Figure 3

This article is protected by copyright. All rights reserved

You might also like