Review Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Jun 18, 2015; 7(11): 1530-1540
Published online Jun 18, 2015. doi: 10.4254/wjh.v7.i11.1530
MicroRNAs dysregulation in hepatocellular carcinoma: Insights in genomic medicine
Iván Lyra-González, Laura E Flores-Fong, Ignacio González-García, David Medina-Preciado, Juan Armendáriz-Borunda, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Instituto de Biología Molecular en Medicina y Terapia Génica, Guadalajara, Jalisco 44281, México
Ignacio González-García, O.P.D. Hospital Civil de Guadalajara “Fray Antonio Alcalde”, Guadalajara, Jalisco 44281, México
David Medina-Preciado, O.P.D. Hospital Civil de Guadalajara “Juan I. Menchaca”, Guadalajara, Jalisco 44281, México
Juan Armendáriz-Borunda, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco 44281, México
Author contributions: All the authors equally contributed to this work.
Conflict-of-interest: Authors have no conflict of interests related to this manuscript. All authors have contributed equally in the realization of this manuscript.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Dr. Juan Armendariz-Borunda, PhD, Head, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Sierra Mojada # 950, Guadalajara, Jalisco 44281, Mexico. armdbo@gmail.com
Telephone: +52-33-10585317 Fax: +52-33-10585318
Received: September 17, 2014
Peer-review started: September 20, 2014
First decision: November 27, 2014
Revised: December 22, 2014
Accepted: May 8, 2015
Article in press: May 11, 2015
Published online: June 18, 2015

Abstract

Hepatocellular carcinoma (HCC) is the leading primary liver cancer and its clinical outcome is still poor. MicroRNAs (miRNAs) have demonstrated an interesting potential to regulate gene expression at post-transcriptional level. Current findings suggest that miRNAs deregulation in cancer is caused by genetic and/or epigenetic, transcriptional and post-transcriptional modifications resulting in abnormal expression and hallmarks of malignant transformation: aberrant cell growth, cell death, differentiation, angiogenesis, invasion and metástasis. The important role of miRNAs in the development and progression of HCC has increased the efforts to understand and develop mechanisms of control overt this single-stranded RNAs. Several studies have analyzed tumoral response to the regulation and control of deregulated miRNAs with good results in vitro and in vivo, proving that targeting aberrant expression of miRNAs is a powerful anticancer therapeutic. Identification of up and/or down regulated miRNAs related to HCC has led to the discovery of new potential application for detection of their presence in the affected organism. MiRNAs represent a relevant new target for diagnosis, prognosis and treatment in a wide variety of pathologic entities, including HCC. This manuscript intends to summarize current knowledge regarding miRNAs and their role in HCC development.

Key Words: Hepatocellular carcinoma, MicroRNAs, Regulation, Therapeutic targets

Core tip: MicroRNAs are implicated in the control of gene expression which enable them a relevant new target for diagnosis, prognosis and treatment in a wide variety of pathologic entities, including hepatocellular carcinoma (HCC). This manuscript represents an attempt to summarize current knowledge regarding miRNAs and their role in HCC development.



INTRODUCTION

Hepatocellular carcinoma (HCC) is the leading primary liver cancer and represents the fifth most common cause of cancer in men, the seventh in women, and is considered the third most frequent cause of cancer-related death worldwide[1]. Almost 85% of new cases occur in developing countries, with highest incidence in areas located in sub-Saharan Africa, east and southeast Asia but also Melanesia and Micronesia/Polynesia; whereas low-incidence areas include northern and Western Europe and North America[1,2]. Nonetheless, clinical outcome of HCC is still poor, which can be attributed to lack of reliable markers for early diagnosis, resistance to treatment, tumor recurrence, and metastasis. Recent evidence suggests a rising incidence of HCC-related deaths in the United States, and during the last two decades, the incidence of HCC in this country has tripled with no difference in 5-year survival rate (12%)[3,4].

HCC develops within an established background of chronic liver disease like cirrhosis due to hepatitis B virus (HBV) and/or HCV, non-alcoholic steatohepatitis, autoimmune hepatitis, iron overload syndromes, diabetes, alcohol abuse, smoking, oral contraceptive use and aflatoxin exposure[5-8].

HCC is believed to be a multistep process, though despite an increasing knowledge of molecular mechanisms inducing hepatocarcinogenesis, poor prognosis of HCC patients reflects the failure to block and reverse the steps of molecular transformation[9,10].

Up to now, alpha-fetoprotein (AFP) along with ultrasounds every 6-12 mo remains as the most commonly used approach to monitoring patients at high risk for HCC[6,11]. Unfortunately the use of both diagnostic tools not only fails to increase detection rates, but also raises false positive uncertainties[12].

Recent studies have demonstrated evidence that anomalous expression of specific miRNAs are implicated in a broad spectrum of human ailments, including rheumatic diseases[13-15], diabetes/insulin resistance[16-18], cardiovascular disease[19-21], renal disease[22] and a wide variety of cancers[23].

Last but not least, the aim of this review is to provide an update in the field of miRNAs and their application in different aspects of HCC.

MIRNAS OVERVIEW AND ITS ROLE IN CANCER DEVELOPMENT

MicroRNAs (miRNAs) are defined as non-coding single-stranded RNAs (ssRNAs) of 19-25 nucleotides in length that are generated from endogenous hairpin-shaped transcripts[24]. MiRNAs were first reported by Lee et al[25], who described a small noncoding RNA encoded by the lin-4 locus associated to the developmental timing of the nematode Caenorhabditis elegans. Since that moment, thousands of miRNAs have been identified in a wide variety of organisms, including mammals and specifically humans. Actually, we know that about 3% of human genes encode miRNAs and more than 1500 miRNA genes have been predicted or experimentally shown to play critical roles in normal cellular functions[26-28].

Up to date, miRNAs have demonstrated an interesting potential to regulate gene expression at post-transcriptional level, binding through partial complementarity to target mRNAs, and mainly leading to mRNA degradation or translation inhibition[29]. Imperfect base pairing between miRNAs and mRNAs is common and enables miRNAs to regulate a broad, but specific set of genes[30].

The first evidence of the involvement of miRNAs in human cancer was reported in chronic lymphocytic leukemia (CLL) patients in 2002, when Calin et al[31] showed miR-16-1 and miR-15a deletion in chromosome 13q14 in more than 59% of CLL patients. Recently, miRNAs alterations have been described in different types of cancer, including CLL, acute promyelocytic leukemia, acute myeloid leukemia, multiple myeloma, monoclonal gammopathy of undetermined significance, non-Hodgkin lymphoma, breast cancer, esophageal cancer, gastric cancer, clear-cell kidney cancer, cervical cancer, and others[23].

Current findings suggest that miRNAs deregulation in cancer is caused by genetic and/or epigenetic, transcriptional, and post-transcriptional modifications resulting in abnormal expression and hallmarks of malignant transformation: aberrant cell growth, cell death, differentiation, angiogenesis, invasion and metastasis[32,33]. This knowledge has established miRNAs as potential diagnostic biomarkers or even as new therapeutic targets in the fight against cancer.

The difficulty of miRNA target prediction and biological validation has been a major obstacle to miRNA research. Experimental identification of miRNAs is difficult to isolate by cloning due to low expression, low stability, tissue specificity and problems in cloning procedures[34].

MIRNAS IN HCC

As discussed before, miRNAs have important functions in cancer development because of their relevant role in regulation of cell proliferation, avoidance of apoptosis (cell perpetuation) and metastasis.

Recently, the identification of up and/or down regulated miRNAs related to HCC has led to the discovery of new potential application for detection of their presence in the affected organism. Up to now, every week appears new evidence of miRNAs with potential effect on carcinogenesis; therefore, in this review we expose the most relevant findings on the field of miRNAs in HCC. To provide an easy comprehension of the data, we have classified our findings based in the up or down regulation status of the most relevant minas implicated in HCC development[35].

Up-regulated miRNAs in HCC

The role of several miRNAs has been studied in other malignances, this is the case of miR-181a which is associated with malignancies such as chronic lymphocytic leukemia and acute myelogenous leukemia[36], and has been linked to improved survival and decrease recurrence in gliomas, where it seems to be an inhibitor of oncogenesis and tumor growth with importance in the development of epithelial cell adhesion molecule+/AFP+ HCC associated with increased metastases and poor survival. Bhattacharya et al[37] analyzed the role of osteopontin (OPN) in HCC, and their findings suggested that OPN confer a prometastatic phenotype to cancer cell lines. Recent findings have described that miR-181 are up-regulated in hepatic stem cell populations and HCC cells with progenitor cell features, implying that miR-181 functions in maintaining an undifferentiated state of hepatic progenitor cells. In this regard, evidence suggests that miR-181 may activate hepatic progenitor cells and HCCs through two cellular signaling pathways: (1) blockage of HCC cell differentiation through inhibition of GATA6 or CDX2, two transcriptional activators regulating hepatocyte differentiation; and (2) activation of Wnt/β-catenin pathway by down-regulating NLK, a Wnt/β-catenin signaling inhibitor[38].

MiR-21 overexpression is found in HCC cells and has been linked to inhibition of apoptosis and promotion of cell proliferation. Connolly et al[39], studied the role of miR-21 in cell invasion and migration, and found that overexpression of this miRNA increases matrix metalloproteinase-9 (MMP-9) activity in multiple cell lines. These findings described the role of MMP-9 expression with invasive and/or metastasic phenotypes of tumors. Other mechanism of metastases identified the role of tumor suppressor RECK, in conjunction with RHOB, in regulating the in vitro metastatic properties, being associated with poor prognosis[39].

MiR-151 is localized within intron 22 of focal adhesion kinase (FAK), which is often overexpressed in human tumors and promotes cancer cell invasion and metastasis. A study carried-out by Ding et al[40] found that suppression of p53 can increase the expression of both FAK and miR-151 simultaneously, suggesting that p53 may be a potential transcriptional regulator for FAK and miR-151 in liver cancer cells. Other description made by this team revealed that RhoGDIA is a direct and functional target for miR-151, which once suppresses RhoGDIA expression activate Rac1, Cdc42 and Rho GTPases, and this inhibitory effect may work synergistically with FAK signaling to promote cell motility and invasion. This situation indicates that it may be a general mechanism for the metastasis of human cancer cells.

Upregulation of miR-191 after hepatocyte injury has been linked with extensive changes in gene expression. The most affected pathways are transforming growth factor beta (TGF-β) and mitogen-activated protein kinases (MAPK) which play a significant role in hepatocarcinogenesis. TGF-β pathway regulates cell proliferation, differentiation, and adhesion. While MEPK signaling pathway is also involved in diverse cellular processes such as cell survival, differentiation, and proliferation[41].

Overexpression of miR-221 is present in almost 71% of HCC and plays an important role in HCC development due to its ability to modulate the expression of the oncogenic proteins c-kit and cyclin-dependent kinase inhibitors CDKN1B/p27 and CDKN1C/p57, promoting cancer cell proliferation. Dysregulation of CDKN1B/p27 exhibits a relevant prognostic significance, being associated with advanced tumor staged, poor survival and recurrence of small HCC. Whereas CDKN1C/p57, has been linked with higher biological aggressiveness, advanced stage, poor differentiation, larger size, portal invasion and high proliferative activity[42]. Other studies showed that miR-221 dysregulation alters G1/S transition inhibitors, where p27 and p21 proteins are frequently down-regulated in HCC, while TGF-β proteins were frequently up-regulated. These alterations lead in loss of control of the transition G1/S in HCC cells, which result in cellular proliferation and metastasis improvement[43]. Furthermore, new evidence suggests a wider role of miRNA in HCC[44], and recently Gramantieri et al[45], described how throughout a pro-apoptotic molecule called Bmf, miR-221 can simultaneously affect proliferation and apoptosis. Bmf is involved in the balance of pro-apoptotic and anti-apoptotic stimuli in Bcl-2/Bcl-xL-induced apoptosis and also seems to follow TGF-β up-regulation[45].

MiR-224 over-expression found in HCC tissues suggests its key role in the malignant phenotype of hepatocarcinoma cells. Recent findings affirmed that miR-224 can modulate cell proliferation and has an important role in cell migration and invasion. Alteration of molecules PAK4 and MMP-9 are considered as the misbalance responsible of the carcinogenic role of miR-224[46].

MiR-183 in the liver acts as negative regulator of programmed cell death 4 (PDCD4) molecule acting at posttranscriptional level which has been found to inhibit activator protein-1 (AP-1) mediated trans-activation and to induce expression of the cyclin-dependent kinase inhibitor p21. MiR-183 up-regulation and subsequent loss of PDCD4 improves cell growing and thereby facilitates cancer development[47]. PDCD4 down-regulation was previously recognized in human colorectal cancer and melanoma[48,49].

Other up-regulated miRNAs related to hepatocarcinogenesis are included in Table 1.

Table 1 Upregulated miRNAs in hepatocellular carcinoma.
MiRNACellular processRef.
MiR-10aEpithelial to mesenchymal transition and metastasis[50]
MiR-130aDrug resistance[51]
MiR-135aMetastasis[52]
MiR-143Metastasis[53]
MiR-155Proliferation and tumorigenesis[54]
MiR-18aProliferation[55]
MiR-181bCell growth, tumorigenesis and metastasis[56]
MiR-182Metastasis[57]
MiR-183Apoptosis[47]
MiR-21Metastasis and drug resistance[39,58,59]
MiR-210Metastasis, apoptosis and proliferation[60,61]
MiR-216aTumorigenesis[62]
MiR-221Apoptosis, proliferation and angiogenesis[42,45,63,64]
MiR-224Metastasis, proliferation and apoptosis[65-67]
MiR-23aGluconeogenesis[68]
MiR-373Cell cycle[69]
MiR-301aMetastasis[70]
MiR-490-3pEpithelial to mesenchymal transition[71]
MiR-519dProliferation, invasion and apoptosis[72]
MiR-550aMetastasis[73]
MiR-590-5pMetastasis and proliferation[74]
MiR-615-5pCell growth and migration[75]
MiR-657Proliferation[76]
MiR-96Proliferation[77,78]
MiR-222Metastasis[79]
Down-regulated miRNAs in HCC

MiR-122 is highly abundant in liver, accounting for 70% of total liver miRNA reported[80-82]. Previous reports had shown its positive regulation of lipid metabolism and disease, but recent knowledge has established an important role of miR-122 in hepatocarcinoma/hepatoma, acting as tumor suppressor gene frequently down-regulated in HCC cell lines and correlated with clinical parameters as etiology, tumor size and differentiation grade. Recent findings suggest that miR-122 inhibits and controls all characteristic properties of cancer cells such as cell cycle, clonogenic survival, anchorage-independent growth, migration, invasion, epithelial-mesenchymal transition and mutagenesis[83-85]. The mechanisms of this dysregulation are unknown, but studies have provided genes and molecules implicated which include ADAM10, Igf1R, SRF, peroxiredoxin 2, members of the septin family like SEPT2 and SEPT9, vimentin, MMP-7, Aldoase A, the muscle isoform of pyruvate kinase (PKM2), and cyclin G1[83-87]. Coulouarn et al[88] showed that repression of miR-122 was characteristic of HCC displaying either a hepatoblast, c-Met or late TGF-β signature; these results showed that HCC cell lines exhibit a more invasive phenotype once decreased miR-122 expression is present. Other study correlated high AFP level with more aggressive properties of HCC. These findings correlated also with lower rates of recurrence-free survival and lower overall survival due to increased expression of CUX1, a direct target of miR-122[89].

Decreased levels of miR-26 in HCC have been associated with poor prognosis and are considered predictive of therapeutic response to interferon-α. Recent studies have reported that animals treated systemically with miR26 presented tumor regression. Recent studies elucidated the role of miR-26 in hepatocyte proliferation confirming that E2 promotes liver cancer cells growth via the E2-ERα pathway and suggested that miR-26 significantly down-regulates ERα preventing hepatoma cell growth, suggesting anti-carcinogenic activities in women[90,91]. Also, miR-26 directly or indirectly regulates expression of a wide variety of genes by down-regulating AFP, PCNA, PR, CEA, nuclear factor-κB and interleukin-6 or increasing P53 and PTEN[90-92].

MiR-34a has been considered a direct transcriptional target of p53 and is commonly reduced or deleted in HCC and other cancers[93]. To date, there are more than 34 proteins altered by miR-34a down-regulation, which include LMNA, ALDH2, MACF1, LOC100129335, GFAP and c-Met as targets of miR-34a with a crucial role in hepatocarcinogenesis[94]. Likewise, down-regulation of miR-34 has shown to down-regulate CyclinD1-CDK6 complex, which is one of the critical positive regulators during G1/S phase transition and a major checkpoint for cell progression. These alterations proved that miR-34a deregulation has the capacity to increase adhesion of tumoral cells to regional lymph nodes improving metastasis[95,96].

Recently, it has been demonstrated that miR-29b is capable of repressing tumor angiogenesis, invasion and metastasis in normal subjects by suppressing MMP-2. Data provided by Fang et al[97], suggest that miR-29b deregulation result in enhanced MMP-2 level in the tumor microenvironment, which in turn activates vascular endothelial growth factor receptor-2 (VEGFR-2) in endothelial cells promoting angiogenesis. Conclusions provided by Fang et al[97], showed inhibitory effects on invasion and metastasis and established MMP-2 as a relevant protein implicated in tumoral growth and metastasis.

MiR-145 forms a double negative feedback loop with key stemness factors OCT4, SOX2, and KLF. And, at the same time, OCT4 binds to the miR-145 promoter and suppresses its expression. Down-regulation of miR-145 in human embryonic stem cells impairs its differentiation and enhances stem cell self-renewal, these findings suggest an important role of miR-145 in carcinogenesis[98]. A study published by Gao et al[99], studied the role of miR-145 in hepatocarcinogenesis and they concluded that down-regulation of miR-145 favors cellular proliferation and migration, suggesting that miR-145 acts as a negative regulator of HCC development.

The analysis of miR-199 down-regulation showed new specific targets like CD44, a member of transmembrane glycoproteins which acts mainly as receptor of hyaluronic acid, being involved in cell-cell interactions, cell adhesion and migration. Studies have demonstrated that inhibition of CD44 enhances apoptosis and improves chemosensitivity, diminishes tumorigenesis and invasion[100]. Interestingly, miR-199 also plays a relevant role in regulation of mammalian target of rapamycin (mTOR) which stands a key role in cell growth, protein translation, metabolism, cell invasion and apoptosis; and c-Met, a proto-oncogene involved in a biological “invasive growth” that result from stimulation of cell motility, invasion, and protection from apoptosis[101,102].

Up-regulation of MKi67 is considered an important risk factor for pathologies in breast, prostate and others cancers like meningiomas, but Hou et al[103] found that higher levels in human HCC cells contribute to malignant phenotype. This study recently published showed that in normal situations, miR-519 suppresses cellular growth by MKi67 due to direct binding of the miRNA to an identified target site in the MKi67 3’-UTR where mutation of this region abolishes this effect.

MiR-152 down-regulation was described as a cause of aberrant DNA methylation by targeting DNMT1, and is inversely correlated with DNMT1 expression in HCC. DNMT1 is necessary and sufficient for maintaining global methylation and aberrant CpG island methylation in human cancer cells contributing to pathogenesis of HCC[104].

Recently, an inverse correlation between miR-338 and smoothened (SMO) expression has been elucidated by Huang et al[105], where miR-338 showed an important role in suppressing HCC metastasis through down-regulating SMO. MMP-9 expression is increased in HCC, correlates with metastasis and advanced tumor stages, and this study has demonstrated that SMO siRNA can abolish MMP-9 expression. These results indicate that miR-338 suppresses the invasiveness of liver cancer through down-regulation of SMO-induced MMP-9 expression[105].

MiR-101 has been shown to be down-regulated in different tumors like breast, lung and pituitary adenoma, but Li et al[106] have demonstrated that its under-expression also has an important role in cell invasion and migration in HCC. This oncogenic activity is attributed to FBJ murine osteosarcoma (FOS), which in normal tissues is negatively regulated by miR-101 at posttranscriptional level via a specific target site within the 3’-UTR. Down-regulation of miR-101 may contribute to the high expression level of FOS protein, which activates the AP-1 family of transcription factors (c-fos and c-jun). Both, c-fos and c-jun can induce epithelial-mesenchymal transition, a hallmark of metastasis and invasive growth associated with loss of cell polarity in epithelial cells. Therefore, according with Li et al[106] regulation of miR-101 could be a potentially suitable candidate for anticancer therapy.

Additional down-regulated miRNAs are included in Table 2.

Table 2 Downregulated microRNAs in hepatocellular carcinoma.
MiRNACellular processRef.
Let-7aApoptosis and proliferation[107,108]
Let-7bApoptosis and proliferation[109]
Let-7cApoptosis, proliferation and cell growth[110-112]
Let-7dApoptosis and proliferation[107]
Let-7f-1Apoptosis and proliferation[107]
Let-7gApoptosis and metastasis[113-115]
MiR-1Proliferation[116]
MiR-34aMetastasis[96]
MiR-101Apoptosis and DNA methylation[110,117,118]
MiR-122Apoptosis, metastasis and angiogenesis[119-122]
MiR-124Proliferation[123]
MiR-125aProliferation, metastasis and metabolism[124-126]
MiR-125bProliferation, metastasis, angiogenesis, apoptosis and histone modification[110,125-127]
MiR-139Metastasis[128,129]
MiR-138Cell cycle[130]
MiR-145Cell growth and tumorigenesis[131]
MiR-195Tumorigenesis, cell cycle and apoptosis[132,133]
MiR-199a-3pDrug resistance and cell growth[101,134]
MiR-199a-5pInvasion and autophagy[135]
MiR-200aProliferation and metastasis[136]
MiR-203Proliferation[137]
MiR-214Cell growth, metastasis and angiogenesis[138,139]
MiR-219-5pProliferation[140]
MiR-223Proliferation[141]
MiR-26a/bCell cycle[142]
MiR-29aProliferation[143]
MiR-34aMetastasis[96,144]
MiR-375Autophagy[145]
MiR-376aApoptosis and proliferation[146]
MiR-449Proliferation and apoptosis[147]
MiR-450aProliferation[148]
MiR-520bCell growth and proliferation[149]
MiR-7Tumorigenesis and metastasis[150]
DISCUSSION

Thus far, more than 800 human miRNAs have been described and speculations about the total number of human miRNAs have exceeded 1000[106]. In human cancer, every type of tumor shows a miRNA profile significantly different compared with normal cells from the same tissue.

Single nucleotide polymorphisms in miRs and their targets have been associated with risk of various cancers because changes in the expression pattern of a gene could therefore influence a person’s risk of illness. Noteworthy, miRs are considered promising prognostic markers of HCC. Some studies have shown that miRs are protected from enzymatic cleavage by RNAses in blood, and therefore their expression profile in serum or plasma could also be utilized as novel diagnostic and prognostic markers[151,152].

Taking into account all this great deal of data, miRNAs issue is one of the most complex topics in oncology due to its wide range of actions as either oncogenes or tumor-suppressors genes in HCC. These facts have led investigators to device two approaches for developing miRNA-based therapies: antagonists and/or mimics[30].

The important role of miRNAs as players in the development and progression of HCC has increased the efforts to understand and develop mechanisms of control overt this ssRNAs. In the last years, several studies have been designed to analyze tumoral response to the regulation and control of deregulated miRNAs with good results in vitro and in vivo, proving that targeting aberrant expression of miRNAs is a powerful anticancer therapeutic[9]. Recent data showed that tumor suppressive miRs expressed in normal liver are down-regulated in tumor tissues during tumorigenesis and metastasis. Hence, a potentially plausible strategy would be to replenish those miRs systemically in HCC patients (miR-181, miR-29, miR-221, miR-122, miR-29, miR-199, etc.) to restore altered pathways balance, and stimulate and/or increase cellular mechanisms to regulate cell proliferation, cell cycle regulation, cell migration and invasion and apoptosis[35].

One of the biggest challenges to translate this knowledge to humans resides that every miRNA may target several mRNAs[78]. This situation empowers selective delivery a crucial issue, which calls for alternate targeted delivery strategy more refined and accurate. The use of viral vectors represents a promising approach[5].

CONCLUSION

MiRNAs are implicated in the control of gene expression which enable them a relevant new target for diagnosis, prognosis and treatment in a wide variety of pathologic entities, including HCC. This manuscript represents an attempt to summarize current knowledge regarding miRNAs and their role in HCC development.

We believe that miRNA is one of the most promising and challenging opportunities to classify and attack cancer. However, translation of knowledge from experimental models to humans remains as a critical point due to the wide and different range of effects caused by each miRNA from cell to cell. Thus, cell-specific delivery most be improved to increase tumoral-specificity and then be considered as a potential therapy in human cancer.

Footnotes

P- Reviewer: Chuang WL, Yang J S- Editor: Tian YL L- Editor: A E- Editor: Liu SQ

References
1.  Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010;127:2893-2917.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11128]  [Cited by in F6Publishing: 11614]  [Article Influence: 893.4]  [Reference Citation Analysis (4)]
2.  Wong CM, Ng IO. Molecular pathogenesis of hepatocellular carcinoma. Liver Int. 2008;28:160-174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 117]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
3.  El-Serag HB. Hepatocellular carcinoma. N Engl J Med. 2011;365:1118-1127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2881]  [Cited by in F6Publishing: 2993]  [Article Influence: 230.2]  [Reference Citation Analysis (0)]
4.  Hamed O, Kimchi ET, Sehmbey M, Gusani NJ, Kaifi JT, Staveley-O’Carroll K. Impact of genetic targets on cancer therapy: hepatocellular cancer. Adv Exp Med Biol. 2013;779:67-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
5.  Lyra-González I, Flores-Fong LE, González-García I, Medina-Preciado D, Armendariz-Borunda J. Adenoviral gene therapy in hepatocellular carcinoma: a review. Hepatol Int. 2013;7:48-58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
6.  Forner A, Llovet JM, Bruix J. Hepatocellular carcinoma. Lancet. 2012;379:1245-1255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3249]  [Cited by in F6Publishing: 3490]  [Article Influence: 290.8]  [Reference Citation Analysis (3)]
7.  Cabibbo G, Maida M, Genco C, Antonucci M, Cammà C. Causes of and prevention strategies for hepatocellular carcinoma. Semin Oncol. 2012;39:374-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
8.  Sanyal AJ, Yoon SK, Lencioni R. The etiology of hepatocellular carcinoma and consequences for treatment. Oncologist. 2010;15 Suppl 4:14-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 325]  [Cited by in F6Publishing: 336]  [Article Influence: 25.8]  [Reference Citation Analysis (0)]
9.  Braconi C, Henry JC, Kogure T, Schmittgen T, Patel T. The role of microRNAs in human liver cancers. Semin Oncol. 2011;38:752-763.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 100]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
10.  Tanaka S, Arii S. Molecular targeted therapies in hepatocellular carcinoma. Semin Oncol. 2012;39:486-492.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 80]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
11.  Ray K. Liver cancer: The promise of new approaches in the management of hepatocellular carcinoma--adding to the toolbox? Nat Rev Gastroenterol Hepatol. 2013;10:195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
12.  Trinchet JC, Chaffaut C, Bourcier V, Degos F, Henrion J, Fontaine H, Roulot D, Mallat A, Hillaire S, Cales P. Ultrasonographic surveillance of hepatocellular carcinoma in cirrhosis: a randomized trial comparing 3- and 6-month periodicities. Hepatology. 2011;54:1987-1997.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 245]  [Cited by in F6Publishing: 259]  [Article Influence: 19.9]  [Reference Citation Analysis (0)]
13.  Alevizos I, Illei GG. MicroRNAs as biomarkers in rheumatic diseases. Nat Rev Rheumatol. 2010;6:391-398.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 164]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
14.  Filková M, Jüngel A, Gay RE, Gay S. MicroRNAs in rheumatoid arthritis: potential role in diagnosis and therapy. BioDrugs. 2012;26:131-141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 79]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
15.  Ceribelli A, Yao B, Dominguez-Gutierrez PR, Nahid MA, Satoh M, Chan EK. MicroRNAs in systemic rheumatic diseases. Arthritis Res Ther. 2011;13:229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 96]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
16.  Dehwah MA, Xu A, Huang Q. MicroRNAs and type 2 diabetes/obesity. J Genet Genomics. 2012;39:11-18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 68]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
17.  Williams MD, Mitchell GM. MicroRNAs in insulin resistance and obesity. Exp Diabetes Res. 2012;2012:484696.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 102]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
18.  Natarajan R, Putta S, Kato M. MicroRNAs and diabetic complications. J Cardiovasc Transl Res. 2012;5:413-422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 75]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
19.  Check JH. Serum CA 125 levels in early pregnancy and subsequent spontaneous abortion. Obstet Gynecol. 1990;76:894-895.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 149]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
20.  van Rooij E, Olson EN. MicroRNA therapeutics for cardiovascular disease: opportunities and obstacles. Nat Rev Drug Discov. 2012;11:860-872.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 460]  [Cited by in F6Publishing: 487]  [Article Influence: 40.6]  [Reference Citation Analysis (0)]
21.  Papageorgiou N, Tousoulis D, Androulakis E, Siasos G, Briasoulis A, Vogiatzi G, Kampoli AM, Tsiamis E, Tentolouris C, Stefanadis C. The role of microRNAs in cardiovascular disease. Curr Med Chem. 2012;19:2605-2610.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
22.  Ho J, Kreidberg JA. The long and short of microRNAs in the kidney. J Am Soc Nephrol. 2012;23:400-404.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
23.  Di Leva G, Croce CM. Roles of small RNAs in tumor formation. Trends Mol Med. 2010;16:257-267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in F6Publishing: 206]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
24.  Kim VN. MicroRNA biogenesis: coordinated cropping and dicing. Nat Rev Mol Cell Biol. 2005;6:376-385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1784]  [Cited by in F6Publishing: 1779]  [Article Influence: 93.6]  [Reference Citation Analysis (0)]
25.  Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75:843-854.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8672]  [Cited by in F6Publishing: 8437]  [Article Influence: 272.2]  [Reference Citation Analysis (0)]
26.  Sassen S, Miska EA, Caldas C. MicroRNA: implications for cancer. Virchows Arch. 2008;452:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 309]  [Cited by in F6Publishing: 325]  [Article Influence: 19.1]  [Reference Citation Analysis (0)]
27.  Xiao ZD, Diao LT, Yang JH, Xu H, Huang MB, Deng YJ, Zhou H, Qu LH. Deciphering the transcriptional regulation of microRNA genes in humans with ACTLocater. Nucleic Acids Res. 2013;41:e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
28.  Holland B, Wong J, Li M, Rasheed S. Identification of human microRNA-like sequences embedded within the protein-encoding genes of the human immunodeficiency virus. PLoS One. 2013;8:e58586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
29.  Iorio MV, Croce CM. MicroRNA dysregulation in cancer: diagnostics, monitoring and therapeutics. A comprehensive review. EMBO Mol Med. 2012;4:143-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1121]  [Cited by in F6Publishing: 1216]  [Article Influence: 101.3]  [Reference Citation Analysis (0)]
30.  Bader AG, Brown D, Stoudemire J, Lammers P. Developing therapeutic microRNAs for cancer. Gene Ther. 2011;18:1121-1126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 222]  [Cited by in F6Publishing: 241]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
31.  Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, Aldler H, Rattan S, Keating M, Rai K. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA. 2002;99:15524-15529.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3675]  [Cited by in F6Publishing: 3636]  [Article Influence: 165.3]  [Reference Citation Analysis (0)]
32.  Benetatos L, Vartholomatos G. Deregulated microRNAs in multiple myeloma. Cancer. 2012;118:878-887.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 39]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
33.  Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19834]  [Cited by in F6Publishing: 18773]  [Article Influence: 782.2]  [Reference Citation Analysis (0)]
34.  ElHefnawi M, Soliman B, Abu-Shahba N, Amer M. An integrative meta-analysis of microRNAs in hepatocellular carcinoma. Genomics Proteomics Bioinformatics. 2013;11:354-367.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
35.  Khare S, Zhang Q, Ibdah JA. Epigenetics of hepatocellular carcinoma: role of microRNA. World J Gastroenterol. 2013;19:5439-5445.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 61]  [Cited by in F6Publishing: 59]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
36.  Marcucci G, Radmacher MD, Maharry K, Mrózek K, Ruppert AS, Paschka P, Vukosavljevic T, Whitman SP, Baldus CD, Langer C. MicroRNA expression in cytogenetically normal acute myeloid leukemia. N Engl J Med. 2008;358:1919-1928.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 355]  [Cited by in F6Publishing: 334]  [Article Influence: 20.9]  [Reference Citation Analysis (0)]
37.  Bhattacharya SD, Garrison J, Guo H, Mi Z, Markovic J, Kim VM, Kuo PC. Micro-RNA-181a regulates osteopontin-dependent metastatic function in hepatocellular cancer cell lines. Surgery. 2010;148:291-297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 40]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
38.  Ji J, Yamashita T, Budhu A, Forgues M, Jia HL, Li C, Deng C, Wauthier E, Reid LM, Ye QH. Identification of microRNA-181 by genome-wide screening as a critical player in EpCAM-positive hepatic cancer stem cells. Hepatology. 2009;50:472-480.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 397]  [Cited by in F6Publishing: 436]  [Article Influence: 29.1]  [Reference Citation Analysis (0)]
39.  Connolly EC, Van Doorslaer K, Rogler LE, Rogler CE. Overexpression of miR-21 promotes an in vitro metastatic phenotype by targeting the tumor suppressor RHOB. Mol Cancer Res. 2010;8:691-700.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 90]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
40.  Ding J, Huang S, Wu S, Zhao Y, Liang L, Yan M, Ge C, Yao J, Chen T, Wan D. Gain of miR-151 on chromosome 8q24.3 facilitates tumour cell migration and spreading through downregulating RhoGDIA. Nat Cell Biol. 2010;12:390-399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 241]  [Cited by in F6Publishing: 258]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
41.  Elyakim E, Sitbon E, Faerman A, Tabak S, Montia E, Belanis L, Dov A, Marcusson EG, Bennett CF, Chajut A. hsa-miR-191 is a candidate oncogene target for hepatocellular carcinoma therapy. Cancer Res. 2010;70:8077-8087.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 130]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
42.  Fornari F, Gramantieri L, Ferracin M, Veronese A, Sabbioni S, Calin GA, Grazi GL, Giovannini C, Croce CM, Bolondi L. MiR-221 controls CDKN1C/p57 and CDKN1B/p27 expression in human hepatocellular carcinoma. Oncogene. 2008;27:5651-5661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
43.  Fu X, Wang Q, Chen J, Huang X, Chen X, Cao L, Tan H, Li W, Zhang L, Bi J. Clinical significance of miR-221 and its inverse correlation with p27Kip¹ in hepatocellular carcinoma. Mol Biol Rep. 2011;38:3029-3035.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
44.  Li J, Wang Y, Yu W, Chen J, Luo J. Expression of serum miR-221 in human hepatocellular carcinoma and its prognostic significance. Biochem Biophys Res Commun. 2011;406:70-73.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 163]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
45.  Gramantieri L, Fornari F, Ferracin M, Veronese A, Sabbioni S, Calin GA, Grazi GL, Croce CM, Bolondi L, Negrini M. MicroRNA-221 targets Bmf in hepatocellular carcinoma and correlates with tumor multifocality. Clin Cancer Res. 2009;15:5073-5081.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 256]  [Article Influence: 17.1]  [Reference Citation Analysis (0)]
46.  Li Q, Wang G, Shan JL, Yang ZX, Wang HZ, Feng J, Zhen JJ, Chen C, Zhang ZM, Xu W. MicroRNA-224 is upregulated in HepG2 cells and involved in cellular migration and invasion. J Gastroenterol Hepatol. 2010;25:164-171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
47.  Li J, Fu H, Xu C, Tie Y, Xing R, Zhu J, Qin Y, Sun Z, Zheng X. miR-183 inhibits TGF-beta1-induced apoptosis by downregulation of PDCD4 expression in human hepatocellular carcinoma cells. BMC Cancer. 2010;10:354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 130]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
48.  Motoyama K, Inoue H, Takatsuno Y, Tanaka F, Mimori K, Uetake H, Sugihara K, Mori M. Over- and under-expressed microRNAs in human colorectal cancer. Int J Oncol. 2009;34:1069-1075.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 172]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
49.  Lin WM, Baker AC, Beroukhim R, Winckler W, Feng W, Marmion JM, Laine E, Greulich H, Tseng H, Gates C. Modeling genomic diversity and tumor dependency in malignant melanoma. Cancer Res. 2008;68:664-673.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 246]  [Cited by in F6Publishing: 245]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
50.  Yan Y, Luo YC, Wan HY, Wang J, Zhang PP, Liu M, Li X, Li S, Tang H. MicroRNA-10a is involved in the metastatic process by regulating Eph tyrosine kinase receptor A4-mediated epithelial-mesenchymal transition and adhesion in hepatoma cells. Hepatology. 2013;57:667-677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 93]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
51.  Xu N, Shen C, Luo Y, Xia L, Xue F, Xia Q, Zhang J. Upregulated miR-130a increases drug resistance by regulating RUNX3 and Wnt signaling in cisplatin-treated HCC cell. Biochem Biophys Res Commun. 2012;425:468-472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 92]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
52.  Liu S, Guo W, Shi J, Li N, Yu X, Xue J, Fu X, Chu K, Lu C, Zhao J. MicroRNA-135a contributes to the development of portal vein tumor thrombus by promoting metastasis in hepatocellular carcinoma. J Hepatol. 2012;56:389-396.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 133]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
53.  Zhang X, Liu S, Hu T, Liu S, He Y, Sun S. Up-regulated microRNA-143 transcribed by nuclear factor kappa B enhances hepatocarcinoma metastasis by repressing fibronectin expression. Hepatology. 2009;50:490-499.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 280]  [Cited by in F6Publishing: 303]  [Article Influence: 20.2]  [Reference Citation Analysis (1)]
54.  Zhang Y, Wei W, Cheng N, Wang K, Li B, Jiang X, Sun S. Hepatitis C virus-induced up-regulation of microRNA-155 promotes hepatocarcinogenesis by activating Wnt signaling. Hepatology. 2012;56:1631-1640.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 235]  [Cited by in F6Publishing: 241]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
55.  Liu WH, Yeh SH, Lu CC, Yu SL, Chen HY, Lin CY, Chen DS, Chen PJ. MicroRNA-18a prevents estrogen receptor-alpha expression, promoting proliferation of hepatocellular carcinoma cells. Gastroenterology. 2009;136:683-693.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 203]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
56.  Wang B, Hsu SH, Majumder S, Kutay H, Huang W, Jacob ST, Ghoshal K. TGFbeta-mediated upregulation of hepatic miR-181b promotes hepatocarcinogenesis by targeting TIMP3. Oncogene. 2010;29:1787-1797.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 287]  [Cited by in F6Publishing: 303]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
57.  Wang J, Li J, Shen J, Wang C, Yang L, Zhang X. MicroRNA-182 downregulates metastasis suppressor 1 and contributes to metastasis of hepatocellular carcinoma. BMC Cancer. 2012;12:227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 123]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
58.  Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST, Patel T. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology. 2007;133:647-658.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2044]  [Cited by in F6Publishing: 2121]  [Article Influence: 124.8]  [Reference Citation Analysis (0)]
59.  Tomimaru Y, Eguchi H, Nagano H, Wada H, Tomokuni A, Kobayashi S, Marubashi S, Takeda Y, Tanemura M, Umeshita K. MicroRNA-21 induces resistance to the anti-tumour effect of interferon-α/5-fluorouracil in hepatocellular carcinoma cells. Br J Cancer. 2010;103:1617-1626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 153]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
60.  Ying Q, Liang L, Guo W, Zha R, Tian Q, Huang S, Yao J, Ding J, Bao M, Ge C. Hypoxia-inducible microRNA-210 augments the metastatic potential of tumor cells by targeting vacuole membrane protein 1 in hepatocellular carcinoma. Hepatology. 2011;54:2064-2075.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 144]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
61.  Yang W, Sun T, Cao J, Liu F, Tian Y, Zhu W. Downregulation of miR-210 expression inhibits proliferation, induces apoptosis and enhances radiosensitivity in hypoxic human hepatoma cells in vitro. Exp Cell Res. 2012;318:944-954.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 90]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
62.  Chen PJ, Yeh SH, Liu WH, Lin CC, Huang HC, Chen CL, Chen DS, Chen PJ. Androgen pathway stimulates microRNA-216a transcription to suppress the tumor suppressor in lung cancer-1 gene in early hepatocarcinogenesis. Hepatology. 2012;56:632-643.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 86]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
63.  Santhekadur PK, Das SK, Gredler R, Chen D, Srivastava J, Robertson C, Baldwin AS, Fisher PB, Sarkar D. Multifunction protein staphylococcal nuclease domain containing 1 (SND1) promotes tumor angiogenesis in human hepatocellular carcinoma through novel pathway that involves nuclear factor κB and miR-221. J Biol Chem. 2012;287:13952-13958.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 110]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
64.  Pineau P, Volinia S, McJunkin K, Marchio A, Battiston C, Terris B, Mazzaferro V, Lowe SW, Croce CM, Dejean A. miR-221 overexpression contributes to liver tumorigenesis. Proc Natl Acad Sci USA. 2010;107:264-269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 542]  [Cited by in F6Publishing: 605]  [Article Influence: 40.3]  [Reference Citation Analysis (0)]
65.  Zhang Y, Takahashi S, Tasaka A, Yoshima T, Ochi H, Chayama K. Involvement of microRNA-224 in cell proliferation, migration, invasion, and anti-apoptosis in hepatocellular carcinoma. J Gastroenterol Hepatol. 2013;28:565-575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 84]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
66.  Wang Y, Toh HC, Chow P, Chung AY, Meyers DJ, Cole PA, Ooi LL, Lee CG. MicroRNA-224 is up-regulated in hepatocellular carcinoma through epigenetic mechanisms. FASEB J. 2012;26:3032-3041.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 88]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
67.  Scisciani C, Vossio S, Guerrieri F, Schinzari V, De Iaco R, D’Onorio de Meo P, Cervello M, Montalto G, Pollicino T, Raimondo G. Transcriptional regulation of miR-224 upregulated in human HCCs by NFκB inflammatory pathways. J Hepatol. 2012;56:855-861.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 110]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
68.  Wang B, Hsu SH, Frankel W, Ghoshal K, Jacob ST. Stat3-mediated activation of microRNA-23a suppresses gluconeogenesis in hepatocellular carcinoma by down-regulating glucose-6-phosphatase and peroxisome proliferator-activated receptor gamma, coactivator 1 alpha. Hepatology. 2012;56:186-197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 175]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
69.  Wu N, Liu X, Xu X, Fan X, Liu M, Li X, Zhong Q, Tang H. MicroRNA-373, a new regulator of protein phosphatase 6, functions as an oncogene in hepatocellular carcinoma. FEBS J. 2011;278:2044-2054.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 52]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
70.  Zhou P, Jiang W, Wu L, Chang R, Wu K, Wang Z. miR-301a is a candidate oncogene that targets the homeobox gene Gax in human hepatocellular carcinoma. Dig Dis Sci. 2012;57:1171-1180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 57]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
71.  Zhang LY, Liu M, Li X, Tang H. miR-490-3p modulates cell growth and epithelial to mesenchymal transition of hepatocellular carcinoma cells by targeting endoplasmic reticulum-Golgi intermediate compartment protein 3 (ERGIC3). J Biol Chem. 2013;288:4035-4047.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 119]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
72.  Fornari F, Milazzo M, Chieco P, Negrini M, Marasco E, Capranico G, Mantovani V, Marinello J, Sabbioni S, Callegari E. In hepatocellular carcinoma miR-519d is up-regulated by p53 and DNA hypomethylation and targets CDKN1A/p21, PTEN, AKT3 and TIMP2. J Pathol. 2012;227:275-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 159]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
73.  Tian Q, Liang L, Ding J, Zha R, Shi H, Wang Q, Huang S, Guo W, Ge C, Chen T. MicroRNA-550a acts as a pro-metastatic gene and directly targets cytoplasmic polyadenylation element-binding protein 4 in hepatocellular carcinoma. PLoS One. 2012;7:e48958.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 44]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
74.  Jiang X, Xiang G, Wang Y, Zhang L, Yang X, Cao L, Peng H, Xue P, Chen D. MicroRNA-590-5p regulates proliferation and invasion in human hepatocellular carcinoma cells by targeting TGF-β RII. Mol Cells. 2012;33:545-551.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 54]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
75.  El Tayebi HM, Hosny KA, Esmat G, Breuhahn K, Abdelaziz AI. miR-615-5p is restrictedly expressed in cirrhotic and cancerous liver tissues and its overexpression alleviates the tumorigenic effects in hepatocellular carcinoma. FEBS Lett. 2012;586:3309-3316.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 46]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
76.  Zhang L, Yang L, Liu X, Chen W, Chang L, Chen L, Loera S, Chu P, Huang WC, Liu YR. MicroRNA-657 promotes tumorigenesis in hepatocellular carcinoma by targeting transducin-like enhancer protein 1 through nuclear factor kappa B pathways. Hepatology. 2013;57:1919-1930.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 54]  [Article Influence: 4.9]  [Reference Citation Analysis (1)]
77.  Xu D, He X, Chang Y, Xu C, Jiang X, Sun S, Lin J. Inhibition of miR-96 expression reduces cell proliferation and clonogenicity of HepG2 hepatoma cells. Oncol Rep. 2013;29:653-661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 56]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
78.  Chen RX, Xia YH, Xue TC, Ye SL. Suppression of microRNA-96 expression inhibits the invasion of hepatocellular carcinoma cells. Mol Med Rep. 2012;5:800-804.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 21]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
79.  Wong QW, Ching AK, Chan AW, Choy KW, To KF, Lai PB, Wong N. MiR-222 overexpression confers cell migratory advantages in hepatocellular carcinoma through enhancing AKT signaling. Clin Cancer Res. 2010;16:867-875.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 195]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
80.  Esau C, Davis S, Murray SF, Yu XX, Pandey SK, Pear M, Watts L, Booten SL, Graham M, McKay R. miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting. Cell Metab. 2006;3:87-98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1612]  [Cited by in F6Publishing: 1588]  [Article Influence: 88.2]  [Reference Citation Analysis (0)]
81.  Lagos-Quintana M, Rauhut R, Yalcin A, Meyer J, Lendeckel W, Tuschl T. Identification of tissue-specific microRNAs from mouse. Curr Biol. 2002;12:735-739.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2502]  [Cited by in F6Publishing: 2504]  [Article Influence: 113.8]  [Reference Citation Analysis (0)]
82.  Kutay H, Bai S, Datta J, Motiwala T, Pogribny I, Frankel W, Jacob ST, Ghoshal K. Downregulation of miR-122 in the rodent and human hepatocellular carcinomas. J Cell Biochem. 2006;99:671-678.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 450]  [Cited by in F6Publishing: 492]  [Article Influence: 28.9]  [Reference Citation Analysis (0)]
83.  Bai S, Nasser MW, Wang B, Hsu SH, Datta J, Kutay H, Yadav A, Nuovo G, Kumar P, Ghoshal K. MicroRNA-122 inhibits tumorigenic properties of hepatocellular carcinoma cells and sensitizes these cells to sorafenib. J Biol Chem. 2009;284:32015-32027.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 383]  [Cited by in F6Publishing: 409]  [Article Influence: 27.3]  [Reference Citation Analysis (0)]
84.  Boutz DR, Collins PJ, Suresh U, Lu M, Ramírez CM, Fernández-Hernando C, Huang Y, Abreu Rde S, Le SY, Shapiro BA. Two-tiered approach identifies a network of cancer and liver disease-related genes regulated by miR-122. J Biol Chem. 2011;286:18066-18078.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 46]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
85.  Diao S, Zhang JF, Wang H, He ML, Lin MC, Chen Y, Kung HF. Proteomic identification of microRNA-122a target proteins in hepatocellular carcinoma. Proteomics. 2010;10:3723-3731.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 44]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
86.  Fornari F, Gramantieri L, Giovannini C, Veronese A, Ferracin M, Sabbioni S, Calin GA, Grazi GL, Croce CM, Tavolari S. MiR-122/cyclin G1 interaction modulates p53 activity and affects doxorubicin sensitivity of human hepatocarcinoma cells. Cancer Res. 2009;69:5761-5767.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 294]  [Cited by in F6Publishing: 325]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
87.  Gramantieri L, Ferracin M, Fornari F, Veronese A, Sabbioni S, Liu CG, Calin GA, Giovannini C, Ferrazzi E, Grazi GL. Cyclin G1 is a target of miR-122a, a microRNA frequently down-regulated in human hepatocellular carcinoma. Cancer Res. 2007;67:6092-6099.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 614]  [Cited by in F6Publishing: 621]  [Article Influence: 36.5]  [Reference Citation Analysis (0)]
88.  Coulouarn C, Factor VM, Andersen JB, Durkin ME, Thorgeirsson SS. Loss of miR-122 expression in liver cancer correlates with suppression of the hepatic phenotype and gain of metastatic properties. Oncogene. 2009;28:3526-3536.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 531]  [Cited by in F6Publishing: 565]  [Article Influence: 37.7]  [Reference Citation Analysis (0)]
89.  Kojima K, Takata A, Vadnais C, Otsuka M, Yoshikawa T, Akanuma M, Kondo Y, Kang YJ, Kishikawa T, Kato N. MicroRNA122 is a key regulator of α-fetoprotein expression and influences the aggressiveness of hepatocellular carcinoma. Nat Commun. 2011;2:338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 89]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
90.  Chen L, Zheng J, Zhang Y, Yang L, Wang J, Ni J, Cui D, Yu C, Cai Z. Tumor-specific expression of microRNA-26a suppresses human hepatocellular carcinoma growth via cyclin-dependent and -independent pathways. Mol Ther. 2011;19:1521-1528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 97]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
91.  Ji J, Shi J, Budhu A, Yu Z, Forgues M, Roessler S, Ambs S, Chen Y, Meltzer PS, Croce CM. MicroRNA expression, survival, and response to interferon in liver cancer. N Engl J Med. 2009;361:1437-1447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 624]  [Cited by in F6Publishing: 638]  [Article Influence: 42.5]  [Reference Citation Analysis (0)]
92.  Kota J, Chivukula RR, O’Donnell KA, Wentzel EA, Montgomery CL, Hwang HW, Chang TC, Vivekanandan P, Torbenson M, Clark KR. Therapeutic microRNA delivery suppresses tumorigenesis in a murine liver cancer model. Cell. 2009;137:1005-1017.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1287]  [Cited by in F6Publishing: 1327]  [Article Influence: 88.5]  [Reference Citation Analysis (0)]
93.  Bommer GT, Gerin I, Feng Y, Kaczorowski AJ, Kuick R, Love RE, Zhai Y, Giordano TJ, Qin ZS, Moore BB. p53-mediated activation of miRNA34 candidate tumor-suppressor genes. Curr Biol. 2007;17:1298-1307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 832]  [Cited by in F6Publishing: 850]  [Article Influence: 50.0]  [Reference Citation Analysis (0)]
94.  Cheng J, Zhou L, Xie QF, Xie HY, Wei XY, Gao F, Xing CY, Xu X, Li LJ, Zheng SS. The impact of miR-34a on protein output in hepatocellular carcinoma HepG2 cells. Proteomics. 2010;10:1557-1572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 62]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
95.  Guo Y, Li S, Qu J, Wang S, Dang Y, Fan J, Yu S, Zhang J. MiR-34a inhibits lymphatic metastasis potential of mouse hepatoma cells. Mol Cell Biochem. 2011;354:275-282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 26]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
96.  Li N, Fu H, Tie Y, Hu Z, Kong W, Wu Y, Zheng X. miR-34a inhibits migration and invasion by down-regulation of c-Met expression in human hepatocellular carcinoma cells. Cancer Lett. 2009;275:44-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 316]  [Cited by in F6Publishing: 351]  [Article Influence: 21.9]  [Reference Citation Analysis (0)]
97.  Fang JH, Zhou HC, Zeng C, Yang J, Liu Y, Huang X, Zhang JP, Guan XY, Zhuang SM. MicroRNA-29b suppresses tumor angiogenesis, invasion, and metastasis by regulating matrix metalloproteinase 2 expression. Hepatology. 2011;54:1729-1740.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 244]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
98.  Xu N, Papagiannakopoulos T, Pan G, Thomson JA, Kosik KS. MicroRNA-145 regulates OCT4, SOX2, and KLF4 and represses pluripotency in human embryonic stem cells. Cell. 2009;137:647-658.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 837]  [Cited by in F6Publishing: 852]  [Article Influence: 56.8]  [Reference Citation Analysis (0)]
99.  Gao P, Wong CC, Tung EK, Lee JM, Wong CM, Ng IO. Deregulation of microRNA expression occurs early and accumulates in early stages of HBV-associated multistep hepatocarcinogenesis. J Hepatol. 2011;54:1177-1184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 119]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
100.  Henry JC, Park JK, Jiang J, Kim JH, Nagorney DM, Roberts LR, Banerjee S, Schmittgen TD. miR-199a-3p targets CD44 and reduces proliferation of CD44 positive hepatocellular carcinoma cell lines. Biochem Biophys Res Commun. 2010;403:120-125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 123]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
101.  Fornari F, Milazzo M, Chieco P, Negrini M, Calin GA, Grazi GL, Pollutri D, Croce CM, Bolondi L, Gramantieri L. MiR-199a-3p regulates mTOR and c-Met to influence the doxorubicin sensitivity of human hepatocarcinoma cells. Cancer Res. 2010;70:5184-5193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 303]  [Cited by in F6Publishing: 328]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
102.  Hou J, Lin L, Zhou W, Wang Z, Ding G, Dong Q, Qin L, Wu X, Zheng Y, Yang Y. Identification of miRNomes in human liver and hepatocellular carcinoma reveals miR-199a/b-3p as therapeutic target for hepatocellular carcinoma. Cancer Cell. 2011;19:232-243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 542]  [Cited by in F6Publishing: 570]  [Article Influence: 43.8]  [Reference Citation Analysis (0)]
103.  Hou YY, Cao WW, Li L, Li SP, Liu T, Wan HY, Liu M, Li X, Tang H. MicroRNA-519d targets MKi67 and suppresses cell growth in the hepatocellular carcinoma cell line QGY-7703. Cancer Lett. 2011;307:182-190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 52]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
104.  Huang J, Wang Y, Guo Y, Sun S. Down-regulated microRNA-152 induces aberrant DNA methylation in hepatitis B virus-related hepatocellular carcinoma by targeting DNA methyltransferase 1. Hepatology. 2010;52:60-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 268]  [Cited by in F6Publishing: 287]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
105.  Huang XH, Chen JS, Wang Q, Chen XL, Wen L, Chen LZ, Bi J, Zhang LJ, Su Q, Zeng WT. miR-338-3p suppresses invasion of liver cancer cell by targeting smoothened. J Pathol. 2011;225:463-472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 100]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
106.  Li S, Fu H, Wang Y, Tie Y, Xing R, Zhu J, Sun Z, Wei L, Zheng X. MicroRNA-101 regulates expression of the v-fos FBJ murine osteosarcoma viral oncogene homolog (FOS) oncogene in human hepatocellular carcinoma. Hepatology. 2009;49:1194-1202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 144]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
107.  Wang Z, Lin S, Li JJ, Xu Z, Yao H, Zhu X, Xie D, Shen Z, Sze J, Li K. MYC protein inhibits transcription of the microRNA cluster MC-let-7a-1~let-7d via noncanonical E-box. J Biol Chem. 2011;286:39703-39714.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 60]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
108.  Tsang WP, Kwok TT. Let-7a microRNA suppresses therapeutics-induced cancer cell death by targeting caspase-3. Apoptosis. 2008;13:1215-1222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 189]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
109.  Di Fazio P, Montalbano R, Neureiter D, Alinger B, Schmidt A, Merkel AL, Quint K, Ocker M. Downregulation of HMGA2 by the pan-deacetylase inhibitor panobinostat is dependent on hsa-let-7b expression in liver cancer cell lines. Exp Cell Res. 2012;318:1832-1843.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 55]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
110.  Au SL, Wong CC, Lee JM, Fan DN, Tsang FH, Ng IO, Wong CM. Enhancer of zeste homolog 2 epigenetically silences multiple tumor suppressor microRNAs to promote liver cancer metastasis. Hepatology. 2012;56:622-631.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 228]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
111.  Zhu XM, Wu LJ, Xu J, Yang R, Wu FS. Let-7c microRNA expression and clinical significance in hepatocellular carcinoma. J Int Med Res. 2011;39:2323-2329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 49]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
112.  Shah YM, Morimura K, Yang Q, Tanabe T, Takagi M, Gonzalez FJ. Peroxisome proliferator-activated receptor alpha regulates a microRNA-mediated signaling cascade responsible for hepatocellular proliferation. Mol Cell Biol. 2007;27:4238-4247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 205]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
113.  Shimizu S, Takehara T, Hikita H, Kodama T, Miyagi T, Hosui A, Tatsumi T, Ishida H, Noda T, Nagano H. The let-7 family of microRNAs inhibits Bcl-xL expression and potentiates sorafenib-induced apoptosis in human hepatocellular carcinoma. J Hepatol. 2010;52:698-704.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 256]  [Cited by in F6Publishing: 263]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
114.  Ji J, Zhao L, Budhu A, Forgues M, Jia HL, Qin LX, Ye QH, Yu J, Shi X, Tang ZY. Let-7g targets collagen type I alpha2 and inhibits cell migration in hepatocellular carcinoma. J Hepatol. 2010;52:690-697.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 167]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
115.  Lan FF, Wang H, Chen YC, Chan CY, Ng SS, Li K, Xie D, He ML, Lin MC, Kung HF. Hsa-let-7g inhibits proliferation of hepatocellular carcinoma cells by downregulation of c-Myc and upregulation of p16(INK4A). Int J Cancer. 2011;128:319-331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 128]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
116.  Li D, Yang P, Li H, Cheng P, Zhang L, Wei D, Su X, Peng J, Gao H, Tan Y. MicroRNA-1 inhibits proliferation of hepatocarcinoma cells by targeting endothelin-1. Life Sci. 2012;91:440-447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 53]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
117.  Su H, Yang JR, Xu T, Huang J, Xu L, Yuan Y, Zhuang SM. MicroRNA-101, down-regulated in hepatocellular carcinoma, promotes apoptosis and suppresses tumorigenicity. Cancer Res. 2009;69:1135-1142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 470]  [Cited by in F6Publishing: 499]  [Article Influence: 33.3]  [Reference Citation Analysis (0)]
118.  Wei X, Xiang T, Ren G, Tan C, Liu R, Xu X, Wu Z. miR-101 is down-regulated by the hepatitis B virus x protein and induces aberrant DNA methylation by targeting DNA methyltransferase 3A. Cell Signal. 2013;25:439-446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 115]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
119.  Lin CJ, Gong HY, Tseng HC, Wang WL, Wu JL. miR-122 targets an anti-apoptotic gene, Bcl-w, in human hepatocellular carcinoma cell lines. Biochem Biophys Res Commun. 2008;375:315-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 189]  [Cited by in F6Publishing: 210]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
120.  Tsai WC, Hsu PW, Lai TC, Chau GY, Lin CW, Chen CM, Lin CD, Liao YL, Wang JL, Chau YP. MicroRNA-122, a tumor suppressor microRNA that regulates intrahepatic metastasis of hepatocellular carcinoma. Hepatology. 2009;49:1571-1582.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 450]  [Cited by in F6Publishing: 465]  [Article Influence: 31.0]  [Reference Citation Analysis (0)]
121.  Reddi HV, Madde P, Milosevic D, Hackbarth JS, Algeciras-Schimnich A, McIver B, Grebe SK, Eberhardt NL. The Putative PAX8/PPARγ Fusion Oncoprotein Exhibits Partial Tumor Suppressor Activity through Up-Regulation of Micro-RNA-122 and Dominant-Negative PPARγ Activity. Genes Cancer. 2011;2:46-55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
122.  Xu J, Zhu X, Wu L, Yang R, Yang Z, Wang Q, Wu F. MicroRNA-122 suppresses cell proliferation and induces cell apoptosis in hepatocellular carcinoma by directly targeting Wnt/β-catenin pathway. Liver Int. 2012;32:752-760.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 171]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
123.  Lang Q, Ling C. MiR-124 suppresses cell proliferation in hepatocellular carcinoma by targeting PIK3CA. Biochem Biophys Res Commun. 2012;426:247-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 108]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
124.  Bi Q, Tang S, Xia L, Du R, Fan R, Gao L, Jin J, Liang S, Chen Z, Xu G. Ectopic expression of MiR-125a inhibits the proliferation and metastasis of hepatocellular carcinoma by targeting MMP11 and VEGF. PLoS One. 2012;7:e40169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 149]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
125.  Kim JK, Noh JH, Jung KH, Eun JW, Bae HJ, Kim MG, Chang YG, Shen Q, Park WS, Lee JY. Sirtuin7 oncogenic potential in human hepatocellular carcinoma and its regulation by the tumor suppressors MiR-125a-5p and MiR-125b. Hepatology. 2013;57:1055-1067.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 227]  [Cited by in F6Publishing: 254]  [Article Influence: 23.1]  [Reference Citation Analysis (0)]
126.  Zhao A, Zeng Q, Xie X, Zhou J, Yue W, Li Y, Pei X. MicroRNA-125b induces cancer cell apoptosis through suppression of Bcl-2 expression. J Genet Genomics. 2012;39:29-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 80]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
127.  Alpini G, Glaser SS, Zhang JP, Francis H, Han Y, Gong J, Stokes A, Francis T, Hughart N, Hubble L. Regulation of placenta growth factor by microRNA-125b in hepatocellular cancer. J Hepatol. 2011;55:1339-1345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 106]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
128.  Wong CC, Wong CM, Tung EK, Au SL, Lee JM, Poon RT, Man K, Ng IO. The microRNA miR-139 suppresses metastasis and progression of hepatocellular carcinoma by down-regulating Rho-kinase 2. Gastroenterology. 2011;140:322-331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 245]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
129.  Fan Q, He M, Deng X, Wu WK, Zhao L, Tang J, Wen G, Sun X, Liu Y. Derepression of c-Fos caused by microRNA-139 down-regulation contributes to the metastasis of human hepatocellular carcinoma. Cell Biochem Funct. 2013;31:319-324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 70]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
130.  Wang W, Zhao LJ, Tan YX, Ren H, Qi ZT. MiR-138 induces cell cycle arrest by targeting cyclin D3 in hepatocellular carcinoma. Carcinogenesis. 2012;33:1113-1120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 163]  [Cited by in F6Publishing: 176]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
131.  Law PT, Ching AK, Chan AW, Wong QW, Wong CK, To KF, Wong N. MiR-145 modulates multiple components of the insulin-like growth factor pathway in hepatocellular carcinoma. Carcinogenesis. 2012;33:1134-1141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 76]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
132.  Xu T, Zhu Y, Xiong Y, Ge YY, Yun JP, Zhuang SM. MicroRNA-195 suppresses tumorigenicity and regulates G1/S transition of human hepatocellular carcinoma cells. Hepatology. 2009;50:113-121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 284]  [Cited by in F6Publishing: 310]  [Article Influence: 20.7]  [Reference Citation Analysis (0)]
133.  Yang X, Yu J, Yin J, Xiang Q, Tang H, Lei X. MiR-195 regulates cell apoptosis of human hepatocellular carcinoma cells by targeting LATS2. Pharmazie. 2012;67:645-651.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
134.  Shatseva T, Lee DY, Deng Z, Yang BB. MicroRNA miR-199a-3p regulates cell proliferation and survival by targeting caveolin-2. J Cell Sci. 2011;124:2826-2836.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 122]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
135.  Shen Q, Cicinnati VR, Zhang X, Iacob S, Weber F, Sotiropoulos GC, Radtke A, Lu M, Paul A, Gerken G. Role of microRNA-199a-5p and discoidin domain receptor 1 in human hepatocellular carcinoma invasion. Mol Cancer. 2010;9:227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 154]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
136.  Yuan JH, Yang F, Chen BF, Lu Z, Huo XS, Zhou WP, Wang F, Sun SH. The histone deacetylase 4/SP1/microrna-200a regulatory network contributes to aberrant histone acetylation in hepatocellular carcinoma. Hepatology. 2011;54:2025-2035.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 139]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
137.  Wei W, Wanjun L, Hui S, Dongyue C, Xinjun Y, Jisheng Z. miR-203 inhibits proliferation of HCC cells by targeting survivin. Cell Biochem Funct. 2013;31:82-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 54]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
138.  Shih TC, Tien YJ, Wen CJ, Yeh TS, Yu MC, Huang CH, Lee YS, Yen TC, Hsieh SY. MicroRNA-214 downregulation contributes to tumor angiogenesis by inducing secretion of the hepatoma-derived growth factor in human hepatoma. J Hepatol. 2012;57:584-591.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 159]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
139.  Xia H, Ooi LL, Hui KM. MiR-214 targets β-catenin pathway to suppress invasion, stem-like traits and recurrence of human hepatocellular carcinoma. PLoS One. 2012;7:e44206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 134]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
140.  Huang N, Lin J, Ruan J, Su N, Qing R, Liu F, He B, Lv C, Zheng D, Luo R. MiR-219-5p inhibits hepatocellular carcinoma cell proliferation by targeting glypican-3. FEBS Lett. 2012;586:884-891.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 68]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
141.  Wong QW, Lung RW, Law PT, Lai PB, Chan KY, To KF, Wong N. MicroRNA-223 is commonly repressed in hepatocellular carcinoma and potentiates expression of Stathmin1. Gastroenterology. 2008;135:257-269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 337]  [Cited by in F6Publishing: 369]  [Article Influence: 23.1]  [Reference Citation Analysis (0)]
142.  Zhu Y, Lu Y, Zhang Q, Liu JJ, Li TJ, Yang JR, Zeng C, Zhuang SM. MicroRNA-26a/b and their host genes cooperate to inhibit the G1/S transition by activating the pRb protein. Nucleic Acids Res. 2012;40:4615-4625.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 169]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
143.  Zhu XC, Dong QZ, Zhang XF, Deng B, Jia HL, Ye QH, Qin LX, Wu XZ. microRNA-29a suppresses cell proliferation by targeting SPARC in hepatocellular carcinoma. Int J Mol Med. 2012;30:1321-1326.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 60]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
144.  Yang P, Li QJ, Feng Y, Zhang Y, Markowitz GJ, Ning S, Deng Y, Zhao J, Jiang S, Yuan Y. TGF-β-miR-34a-CCL22 signaling-induced Treg cell recruitment promotes venous metastases of HBV-positive hepatocellular carcinoma. Cancer Cell. 2012;22:291-303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 394]  [Cited by in F6Publishing: 419]  [Article Influence: 34.9]  [Reference Citation Analysis (0)]
145.  Chang Y, Yan W, He X, Zhang L, Li C, Huang H, Nace G, Geller DA, Lin J, Tsung A. miR-375 inhibits autophagy and reduces viability of hepatocellular carcinoma cells under hypoxic conditions. Gastroenterology. 2012;143:177-187.e8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 217]  [Article Influence: 18.1]  [Reference Citation Analysis (0)]
146.  Zheng Y, Yin L, Chen H, Yang S, Pan C, Lu S, Miao M, Jiao B. miR-376a suppresses proliferation and induces apoptosis in hepatocellular carcinoma. FEBS Lett. 2012;586:2396-2403.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 57]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
147.  Buurman R, Gürlevik E, Schäffer V, Eilers M, Sandbothe M, Kreipe H, Wilkens L, Schlegelberger B, Kühnel F, Skawran B. Histone deacetylases activate hepatocyte growth factor signaling by repressing microRNA-449 in hepatocellular carcinoma cells. Gastroenterology. 2012;143:811-20.e1-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 156]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
148.  Weng Z, Wang D, Zhao W, Song M, You F, Yang L, Chen L. microRNA-450a targets DNA methyltransferase 3a in hepatocellular carcinoma. Exp Ther Med. 2011;2:951-955.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
149.  Zhang W, Kong G, Zhang J, Wang T, Ye L, Zhang X. MicroRNA-520b inhibits growth of hepatoma cells by targeting MEKK2 and cyclin D1. PLoS One. 2012;7:e31450.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 55]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
150.  Fang Y, Xue JL, Shen Q, Chen J, Tian L. MicroRNA-7 inhibits tumor growth and metastasis by targeting the phosphoinositide 3-kinase/Akt pathway in hepatocellular carcinoma. Hepatology. 2012;55:1852-1862.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 318]  [Cited by in F6Publishing: 329]  [Article Influence: 27.4]  [Reference Citation Analysis (0)]
151.  Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat Rev Cancer. 2006;6:857-866.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5705]  [Cited by in F6Publishing: 5849]  [Article Influence: 324.9]  [Reference Citation Analysis (0)]
152.  Gilad S, Meiri E, Yogev Y, Benjamin S, Lebanony D, Yerushalmi N, Benjamin H, Kushnir M, Cholakh H, Melamed N. Serum microRNAs are promising novel biomarkers. PLoS One. 2008;3:e3148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 996]  [Cited by in F6Publishing: 1042]  [Article Influence: 65.1]  [Reference Citation Analysis (0)]