Skip to main content
World Journal of Hepatology logoLink to World Journal of Hepatology
. 2015 Jan 27;7(1):1–6. doi: 10.4254/wjh.v7.i1.1

Diagnostic and therapeutic application of noncoding RNAs for hepatocellular carcinoma

Chikako Shibata 1,2, Motoyuki Otsuka 1,2, Takahiro Kishikawa 1,2, Motoko Ohno 1,2, Takeshi Yoshikawa 1,2, Akemi Takata 1,2, Kazuhiko Koike 1,2
PMCID: PMC4295186  PMID: 25624991

Abstract

MicroRNAs (miRNAs) are small, noncoding RNA molecules that regulate gene expression posttranscriptionally, targeting thousands of messenger RNAs. Long noncoding RNAs (lncRNAs), another class of noncoding RNAs, have been determined to be also involved in transcription regulation and translation of target genes. Since deregulated expression levels or functions of miRNAs and lncRNAs in hepatocellular carcinoma (HCC) are frequently observed, clinical use of noncoding RNAs for novel diagnostic and therapeutic applications in the management of HCCs is highly and emergently expected. Here, we summarize recent findings regarding deregulated miRNAs and lncRNAs for their potential clinical use as diagnostic and prognostic biomarkers of HCC. Specifically, we emphasize the deregulated expression levels of such noncoding RNAs in patients’ sera as noninvasive biomarkers, a field that requires urgent improvement in the clinical surveillance of HCC. Since nucleotide-based strategies are being applied to clinical therapeutics, we further summarize clinical and preclinical trials using oligonucleotides involving the use of miRNAs and small interfering RNAs against HCC as novel therapeutics. Finally, we discuss current open questions, which must be clarified in the near future for realistic clinical applications of these new strategies.

Keywords: MicroRNA, Long noncoding RNA, Hepatocellular carcinoma, Clinical trials, Biomarker


Core tip: In this review, we summarize the latest findings on deregulated microRNAs (miRNAs) and long noncoding RNAs in hepatocellular carcinomas (HCCs) with a focus on their clinical use as novel diagnostic and prognostic biomarkers. In addition, we summarize the current status of clinical and preclinical oligonucleotide therapies including miRNAs and small interfering RNAs as novel HCC therapeutics. This review will enable the readers to understand the current status of clinical applications and knowledge of noncoding RNAs in HCC management.

INTRODUCTION

Noncoding RNAs contain multiple classes of RNAs that are not transcribed into proteins. While most noncoding RNAs studied to date are microRNAs (miRNAs), many noncoding RNAs with various lengths have also been reported.

MiRNAs are short, single-stranded RNAs that are expressed in most organisms[1-3]. Through gene expression regulation at a posttranscriptional level, miRNAs are involved in various physiological and pathological processes[4,5]. Since the discovery of miRNA lin-4 in Caenorhabditis elegans[6,7], as of August 2014, 1881 miRNA precursors and 2588 mature miRNA sequences in humans are deposited in miRBase, a miRNA database by the Sanger Institute[8]. MiRNAs are dysregulated in nearly all types of cancer[9,10], and specific signatures of aberrantly expressed miRNAs in specific cancers may have diagnostic and therapeutic implications[11,12].

Long noncoding RNAs (lncRNAs) also play crucial roles in transcription and translation[13,14]. Similar to miRNAs, their dysregulation is also associated with human cancers[15]. One of the most well-studied lncRNAs is the HOX transcript antisense intergenic RNA (HOTAIR). Class I homeobox genes (HOX in humans) encode 39 transcriptional factors initially described as master regulators of embryonic development[16] and display a unique gene network organization. HOTAIR, a 2.2-kb-long RNA residing within the HOXC locus, was initially described in breast cancer tissues, where it is highly expressed[17]. In addition to HOTAIR, many other lncRNAs are dysregulated in cancer tissues. Thus, lncRNAs may also be candidates for biomarker discovery and therapeutic applications in hepatocellular carcinomas (HCCs)[18].

In contrast to miRNAs and lncRNAs, short interfering RNAs (siRNAs) are double-stranded RNAs that degrade mRNAs through perfect matches with their target sequences. Although human telomerase reverse transcriptase was recently found to function as an RNA-dependent RNA polymerase and contribute to RNA silencing[19], its activities are not dominant in mammals. Additionally, endogenously produced siRNAs may play functional roles under limited circumstances in humans[20]. However, the exogenous application of synthesized siRNAs is an attractive method that could be used to intervene in crucial gene expression under pathological conditions, including cancers[21].

HCC is the third leading cause of cancer-related mortality worldwide[22]. Although advances have been made in early detection and interventional therapies, a continuing need exists to develop novel approaches for the management of advanced HCC[23]. While many reports have described deregulated expression levels or functions of miRNAs and lncRNAs in HCCs, we will focus on the potential clinical use of noncoding RNAs in the very near future for novel diagnostic and therapeutic applications in the management of HCCs.

NONCODING RNAS AS BIOMARKERS FOR HCC

Deregulated expression levels of noncoding RNAs in HCC tissues

Although several published reports have described deregulated expression levels of miRNAs and lncRNAs in HCC tissues[18,24,25], the data thus far vary greatly. The differences may be because of several reasons, including the use of different techniques or samples as controls, normal liver tissues vs nonneoplastic tissues around tumors, background livers with various fibrosis staging, inflammation activities, or etiologies, such as hepatitis B, hepatitis C, or steatohepatitis, as well as the age or sex of the tissue-derived patients; any of these factors may cause the differential expression status of miRNAs. Regardless of these limitations, the plenty data about dysregulated miRNAs in HCCs suggests that noncoding RNAs play crucial roles in hepatocarcinogenesis[24].

Deregulated expression of noncoding RNAs in HCC as prognostic/diagnostic markers

Deregulated expression levels of noncoding RNAs in HCC tissues that may be clinically useful as prognostic/diagnostic markers will be described herein. The landmark paper that initially addressed this issue focused on miR26 expression levels in HCC tissues and was published in the New England Journal of Medicine[26]. In this study, HCC showed frequently reduced levels of miR26, and patients exhibited low miR26 expression with a shorter overall survival but a better response to interferon therapy, indicating that miRNA expression status is associated with survival and response to therapy.

Expression levels of miRNAs have tissue specificities. In the liver, miR122, miR192, and miR199a/b-3p are highly expressed miRNAs of all mRNAs in the liver[27]. The role of miR122 loss in hepatocarcinogenesis was confirmed in a mouse model[28,29], and its expression is decreased in HCCs, especially non-viral HCCs[27]. Decreased expression of miR122 is also linked with poor prognosis of HCC[30]. Although miR192 was not deregulated in HCCs in previous studies, miR199a/b-3p is frequently decreased in HCCs[27]. In contrast, miR21, whose expression is increased when rat hepatectomy[31], is upregulated as an onco-miRNA, resulting in the promotion of HCC[32]. MiR21 expression in HCC tissues confers resistance to the antitumor effect of interferon-α and 5FU combination therapy[33].

Similar to miRNAs, expression levels of lncRNAs are also dysregulated in HCC tissues[18]. Among them, HOTAIR is overexpressed in HCC tissues and may confer chemoresistance[34]. Metastasis-associated lung adenocarcinoma transcript 1, which was initially discovered as an lncRNA associated with metastasis[35], is also upregulated in HCC tissues and may be useful as a biomarker for tumor recurrence. Recently, HOXA transcript at the distal tip (HOTTIP) was discovered to be located in physical contiguity with the HOXA13 gene and upregulated in HCC tissues, and this was also associated with metastasis formation and poor patient survival[36]. These results show the functional importance of lncRNA dysregulation in HCC tissues and indicate their possible use as novel prognostic and diagnostic biomarkers.

Noncoding RNAs in the sera of patients with HCC as diagnostic markers

Although α-fetoprotein (AFP), AFP-L3, and des-gamma-carboxy prothrombin are useful noninvasive biomarkers for HCC surveillance[37], novel and sensitive biomarkers that can detect early HCC are needed. The identification of tumor-specific alterations in circulating nucleic acids of patients with cancer as noninvasive methods of cancer diagnosis is encouraging[38]. Although RNAs are generally considered unstable, they are actually quite stable and readily detected in patient serum and plasma. Microarrays, polymerase chain reaction methods, and next-generation sequencing technologies are generally utilized to detect circulating noncoding RNAs.

Although many reports have described circulating miRNA levels in patients with HCC, only a few tests have been reproducible. For example, data regarding upregulation of circulating miR21, miR222, and miR223 in patients with HCC are inconsistent[32,33,38-44]. Highly upregulated in liver cancer, a 1.6-kb lncRNA, is also upregulated in HCC tissues[45-47] and is detected in the plasma of patients with HCC[18,48]. Although these results are encouraging, more work is needed to make the usability of circulating noncoding RNAs as novel biomarkers more reliable (Table 1). Specificity and sensitivity, as well as methods to quantitate small amounts of RNAs in sera with high reproducibility and the universal control to adjust the obtained data from differing times and samples, need to be urgently determined[49].

Table 1.

Representative noncoding RNAs in sera for Hepatocellular carcinoma diagnosis

MiRNA Expression levels in HCC Possible targets Ref.
MiR21 Upregulated PTEN, AKT, C/EBPb [32,39,58]
MiR222 Upregulated PP2A, p27, DDIT4 [42,43,59]
MiR223 Upregulated Stathmin [44]
HULC Upregulated IGF2BP1 [45-47]

HCC: Hepatocellular carcinoma; HULC: Highly up-regulated in liver cancer; PTEN: Phosphatase and tensin-like protein; AKT: V-akt murine thymoma viral oncogene homolog; C/EBPb: CCAAT/enhancer-binding protein beta; PP2A: Protein phosphatase 2A; IGF2BP1: Insulin-like growth factor 2 MRNA binding protein 1.

NONCODING RNAS AS NOVEL THERAPEUTICS AGAINST HCC

Ongoing clinical trials

Mounting evidence suggests that noncoding RNAs are frequently dysregulated in HCCs and possibly involved in oncogenesis and may therefore provide novel molecular targets as a therapeutic intervention. However, due to the complexity associated with pleiotropic miRNA functions and lncRNAs, the number of clinical trials is presently limited[50]. The leading nucleotide-targeting therapy, Miravirsen, an LNA-based anti-miR122 against hepatitis C virus replication, has been successful in a Phase IIa study[51]. In addition, MRX34, a liposome-formulated miR-34 mimic developed by Mirna Therapeutics, produced complete HCC regression in mouse models[52], and a Phase I study is currently recruiting patients with advanced liver cancer for HCC therapeutic intervention (NCT01829971).

While siRNAs are not endogenous noncoding RNAs, they can be described as noncoding RNAs that have been tried as novel therapeutics against HCC. ALN-VSP (Alnylam Pharmaceuticals), an RNAi therapeutic targeting vascular endothelial growth factor and kinesin spindle protein, has been shown to be well tolerated in Phase I studies (NCT008822180 and NCT01158079) for the treatment of primary and metastatic liver cancer. The results demonstrated disease control lasting more than 6 mo in the majority of patients, including a complete response in a patient with endometrial cancer who had multiple liver metastases. TMK-polo-like kinase 1 (PLK1) (Tekmira Pharmaceuticals), an RNAi targeting PLK1, is also under a Phase I/II trial (NCT01437007). Early results show that TKM-PLK is well tolerated and demonstrates clinical benefits. Although primary results from these potential therapeutics are encouraging, the benefits and unexpected side effects need to be determined, especially under long-term use.

Preclinical trials

Anti-miR21 and anti-miR221 are under development for clinical use (Regulus Therapeutics). MiR21 is one of the most validated microRNA targets, with numerous scientific publications suggesting that miR21 plays an important role in the initiation and progression of cancers, including liver cancer[32,53,54]. Similarly, miR221 has been identified to be upregulated in multiple cancers including liver cancer[54-56]. Anti-miR21 and anti-miR221 prolonged survival time in a preclinical mouse model that genetically develops HCC. An miR7 mimic is also under development (MiReven). Mir7 targets the phosphoinositide 3-kinase (PI3K) pathway and decreases tumor growth both in vitro and in vivo[57]. These results are summarized in Table 2.

Table 2.

Representative noncoding RNAs under clinical and preclinical trials for hepatocellular carcinoma therapeutics

Target Name Content Vendor Current status
MiR34 MRX34 Liposome-formulated miR-34 mimic Mirna Therapeutics Phase I
VEGF/KSP ALN-VSP RNAi targeting VEGF/KSP Alnylam Pharmaceuticals Phase I
PLK1 TMK-PLK1 RNAi targeting PLK1 Tekmira Pharmaceuticals Phase I/II
MiR21 Anti-miR21 Antisense against miR21 Regulus Therapeutics Preclinical
MiR221 Anti-miR221 Antisense against miR221 Regulus Therapeutics Preclinical
MiR7 MiR7 mimic MiR7 mimic MiReven Preclinical

VEGF: Vascular endothelial growth factor; KSP: Kidney-specific cadherin; PLK1: Polo-like kinase 1.

CHALLENGES FOR BETTER CLINICAL TRANSLATION

Several other miRNAs, including lncRNAs, which are dysregulated in HCCs, can be attractive therapeutic targets by RNA mimics, antisense RNA, or siRNA. In fact, many publications have reported their efficacy. However, obstacles remain to be addressed[24]: (1) The more reproducibility of the results should be achieved to make the data more reliable; (2) Identification of driver miRNAs in oncogenesis is important to develop therapeutics targeting such miRNAs, although we may be able to use passive miRNAs as prognostic and diagnostic bio-markers; and (3) The delivery methods of oligonucleotides into specific tissues with improved oligonucleotide modification, and safety need to be seriously considered for utilizing miRNAs in clinical applications. Because miRNAs generally target multiple mRNAs, unexpected outcomes, “off-target effects,” may occur, even when targeting a single miRNA.

More research to solve these issues is definitely needed for the improved translational application utilizing the data about miRNAs in HCCs.

CONCLUSION

The discovery of miRNAs and lncRNAs has opened up new possibilities for novel diagnostic and therapeutic tools against HCCs. However, several important issues remain to be resolved. We must conduct continuous research to develop innovative and useful applications of the miRNA data in the clinical management of HCCs.

Footnotes

P- Reviewer: Tarazov PG, Vespasiani-Gentilucci U S- Editor: Gong XM L- Editor: A E- Editor: Liu SQ

Supported by Grants-in-Aid from the Ministry of Education, Culture, Sports, Science and Technology, Japan, Nos. #25293076, #26860492, #25860520, and #24390183 (to Otsuka M, Kishikawa T, Yoshikawa T and Koike K); by Health Sciences Research Grants of The Ministry of Health, Labour and Welfare of Japan (to Koike K); by grants from the Japanese Society of Gastroenterology, Okinaka Memorial Institute for Medical Research, and Honjo International Scholarship Foundation (to Otsuka M); by a grant from the Mishima Kaiun Memorial Foundation (to Ohno M)

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/

Peer-review started: August 20, 2014

First decision: September 16, 2014

Article in press: November 19, 2014

References

  • 1.Ebert MS, Sharp PA. Roles for microRNAs in conferring robustness to biological processes. Cell. 2012;149:515–524. doi: 10.1016/j.cell.2012.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Cech TR, Steitz JA. The noncoding RNA revolution-trashing old rules to forge new ones. Cell. 2014;157:77–94. doi: 10.1016/j.cell.2014.03.008. [DOI] [PubMed] [Google Scholar]
  • 3.Mendell JT, Olson EN. MicroRNAs in stress signaling and human disease. Cell. 2012;148:1172–1187. doi: 10.1016/j.cell.2012.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ambros V. The functions of animal microRNAs. Nature. 2004;431:350–355. doi: 10.1038/nature02871. [DOI] [PubMed] [Google Scholar]
  • 5.Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–297. doi: 10.1016/s0092-8674(04)00045-5. [DOI] [PubMed] [Google Scholar]
  • 6.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. doi: 10.1016/0092-8674(93)90529-y. [DOI] [PubMed] [Google Scholar]
  • 7.Wightman B, Ha I, Ruvkun G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell. 1993;75:855–862. doi: 10.1016/0092-8674(93)90530-4. [DOI] [PubMed] [Google Scholar]
  • 8.Kozomara A, Griffiths-Jones S. miRBase: integrating microRNA annotation and deep-sequencing data. Nucleic Acids Res. 2011;39:D152–D157. doi: 10.1093/nar/gkq1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, et al. MicroRNA expression profiles classify human cancers. Nature. 2005;435:834–838. doi: 10.1038/nature03702. [DOI] [PubMed] [Google Scholar]
  • 10.Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C, Ferracin M, et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA. 2006;103:2257–2261. doi: 10.1073/pnas.0510565103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Esteller M. Non-coding RNAs in human disease. Nat Rev Genet. 2011;12:861–874. doi: 10.1038/nrg3074. [DOI] [PubMed] [Google Scholar]
  • 12.Ling H, Fabbri M, Calin GA. MicroRNAs and other non-coding RNAs as targets for anticancer drug development. Nat Rev Drug Discov. 2013;12:847–865. doi: 10.1038/nrd4140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wilusz JE, Sunwoo H, Spector DL. Long noncoding RNAs: functional surprises from the RNA world. Genes Dev. 2009;23:1494–1504. doi: 10.1101/gad.1800909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kung JT, Colognori D, Lee JT. Long noncoding RNAs: past, present, and future. Genetics. 2013;193:651–669. doi: 10.1534/genetics.112.146704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Gutschner T, Diederichs S. The hallmarks of cancer: a long non-coding RNA point of view. RNA Biol. 2012;9:703–719. doi: 10.4161/rna.20481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Graham A, Papalopulu N, Krumlauf R. The murine and Drosophila homeobox gene complexes have common features of organization and expression. Cell. 1989;57:367–378. doi: 10.1016/0092-8674(89)90912-4. [DOI] [PubMed] [Google Scholar]
  • 17.Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 2010;464:1071–1076. doi: 10.1038/nature08975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.He Y, Meng XM, Huang C, Wu BM, Zhang L, Lv XW, Li J. Long noncoding RNAs: Novel insights into hepatocelluar carcinoma. Cancer Lett. 2014;344:20–27. doi: 10.1016/j.canlet.2013.10.021. [DOI] [PubMed] [Google Scholar]
  • 19.Maida Y, Yasukawa M, Furuuchi M, Lassmann T, Possemato R, Okamoto N, Kasim V, Hayashizaki Y, Hahn WC, Masutomi K. An RNA-dependent RNA polymerase formed by TERT and the RMRP RNA. Nature. 2009;461:230–235. doi: 10.1038/nature08283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Watanabe T, Totoki Y, Toyoda A, Kaneda M, Kuramochi-Miyagawa S, Obata Y, Chiba H, Kohara Y, Kono T, Nakano T, et al. Endogenous siRNAs from naturally formed dsRNAs regulate transcripts in mouse oocytes. Nature. 2008;453:539–543. doi: 10.1038/nature06908. [DOI] [PubMed] [Google Scholar]
  • 21.Sehgal A, Vaishnaw A, Fitzgerald K. Liver as a target for oligonucleotide therapeutics. J Hepatol. 2013;59:1354–1359. doi: 10.1016/j.jhep.2013.05.045. [DOI] [PubMed] [Google Scholar]
  • 22.Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin. 2005;55:74–108. doi: 10.3322/canjclin.55.2.74. [DOI] [PubMed] [Google Scholar]
  • 23.Greten TF, Korangy F, Manns MP, Malek NP. Molecular therapy for the treatment of hepatocellular carcinoma. Br J Cancer. 2009;100:19–23. doi: 10.1038/sj.bjc.6604784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Otsuka M, Kishikawa T, Yoshikawa T, Ohno M, Takata A, Shibata C, Koike K. The role of microRNAs in hepatocarcinogenesis: current knowledge and future prospects. J Gastroenterol. 2014;49:173–184. doi: 10.1007/s00535-013-0909-8. [DOI] [PubMed] [Google Scholar]
  • 25.Petrelli A, Perra A, Cora D, Sulas P, Menegon S, Manca C, Migliore C, Kowalik MA, Ledda-Columbano GM, Giordano S, et al. MicroRNA/gene profiling unveils early molecular changes and nuclear factor erythroid related factor 2 (NRF2) activation in a rat model recapitulating human hepatocellular carcinoma (HCC) Hepatology. 2014;59:228–241. doi: 10.1002/hep.26616. [DOI] [PubMed] [Google Scholar]
  • 26.Ji J, Shi J, Budhu A, Yu Z, Forgues M, Roessler S, Ambs S, Chen Y, Meltzer PS, Croce CM, et al. MicroRNA expression, survival, and response to interferon in liver cancer. N Engl J Med. 2009;361:1437–1447. doi: 10.1056/NEJMoa0901282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hou J, Lin L, Zhou W, Wang Z, Ding G, Dong Q, Qin L, Wu X, Zheng Y, Yang Y, et al. 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. doi: 10.1016/j.ccr.2011.01.001. [DOI] [PubMed] [Google Scholar]
  • 28.Hsu SH, Wang B, Kota J, Yu J, Costinean S, Kutay H, Yu L, Bai S, La Perle K, Chivukula RR, et al. Essential metabolic, anti-inflammatory, and anti-tumorigenic functions of miR-122 in liver. J Clin Invest. 2012;122:2871–2883. doi: 10.1172/JCI63539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Tsai WC, Hsu SD, Hsu CS, Lai TC, Chen SJ, Shen R, Huang Y, Chen HC, Lee CH, Tsai TF, et al. MicroRNA-122 plays a critical role in liver homeostasis and hepatocarcinogenesis. J Clin Invest. 2012;122:2884–2897. doi: 10.1172/JCI63455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kojima K, Takata A, Vadnais C, Otsuka M, Yoshikawa T, Akanuma M, Kondo Y, Kang YJ, Kishikawa T, Kato N, et al. MicroRNA122 is a key regulator of α-fetoprotein expression and influences the aggressiveness of hepatocellular carcinoma. Nat Commun. 2011;2:338. doi: 10.1038/ncomms1345. [DOI] [PubMed] [Google Scholar]
  • 31.Castro RE, Ferreira DM, Zhang X, Borralho PM, Sarver AL, Zeng Y, Steer CJ, Kren BT, Rodrigues CM. Identification of microRNAs during rat liver regeneration after partial hepatectomy and modulation by ursodeoxycholic acid. Am J Physiol Gastrointest Liver Physiol. 2010;299:G887–G897. doi: 10.1152/ajpgi.00216.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.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. doi: 10.1053/j.gastro.2007.05.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Tomimaru Y, Eguchi H, Nagano H, Wada H, Tomokuni A, Kobayashi S, Marubashi S, Takeda Y, Tanemura M, Umeshita K, et al. 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. doi: 10.1038/sj.bjc.6605958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Yang F, Zhang L, Huo XS, Yuan JH, Xu D, Yuan SX, Zhu N, Zhou WP, Yang GS, Wang YZ, et al. Long noncoding RNA high expression in hepatocellular carcinoma facilitates tumor growth through enhancer of zeste homolog 2 in humans. Hepatology. 2011;54:1679–1689. doi: 10.1002/hep.24563. [DOI] [PubMed] [Google Scholar]
  • 35.Ji P, Diederichs S, Wang W, Böing S, Metzger R, Schneider PM, Tidow N, Brandt B, Buerger H, Bulk E, et al. MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene. 2003;22:8031–8041. doi: 10.1038/sj.onc.1206928. [DOI] [PubMed] [Google Scholar]
  • 36.Quagliata L, Matter MS, Piscuoglio S, Arabi L, Ruiz C, Procino A, Kovac M, Moretti F, Makowska Z, Boldanova T, et al. Long noncoding RNA HOTTIP/HOXA13 expression is associated with disease progression and predicts outcome in hepatocellular carcinoma patients. Hepatology. 2014;59:911–923. doi: 10.1002/hep.26740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Huang J, Zeng Y. Current clinical uses of the biomarkers for hepatocellular carcinoma. Drug Discov Ther. 2014;8:98–99. doi: 10.5582/ddt.8.98. [DOI] [PubMed] [Google Scholar]
  • 38.Qu KZ, Zhang K, Li H, Afdhal NH, Albitar M. Circulating microRNAs as biomarkers for hepatocellular carcinoma. J Clin Gastroenterol. 2011;45:355–360. doi: 10.1097/MCG.0b013e3181f18ac2. [DOI] [PubMed] [Google Scholar]
  • 39.Xu J, Wu C, Che X, Wang L, Yu D, Zhang T, Huang L, Li H, Tan W, Wang C, et al. Circulating microRNAs, miR-21, miR-122, and miR-223, in patients with hepatocellular carcinoma or chronic hepatitis. Mol Carcinog. 2011;50:136–142. doi: 10.1002/mc.20712. [DOI] [PubMed] [Google Scholar]
  • 40.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. doi: 10.1016/j.bbrc.2011.01.111. [DOI] [PubMed] [Google Scholar]
  • 41.Tomimaru Y, Eguchi H, Nagano H, Wada H, Kobayashi S, Marubashi S, Tanemura M, Tomokuni A, Takemasa I, Umeshita K, et al. Circulating microRNA-21 as a novel biomarker for hepatocellular carcinoma. J Hepatol. 2012;56:167–175. doi: 10.1016/j.jhep.2011.04.026. [DOI] [PubMed] [Google Scholar]
  • 42.Qi P, Cheng SQ, Wang H, Li N, Chen YF, Gao CF. Serum microRNAs as biomarkers for hepatocellular carcinoma in Chinese patients with chronic hepatitis B virus infection. PLoS One. 2011;6:e28486. doi: 10.1371/journal.pone.0028486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.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. doi: 10.1158/1078-0432.CCR-09-1840. [DOI] [PubMed] [Google Scholar]
  • 44.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. doi: 10.1053/j.gastro.2008.04.003. [DOI] [PubMed] [Google Scholar]
  • 45.Hämmerle M, Gutschner T, Uckelmann H, Ozgur S, Fiskin E, Gross M, Skawran B, Geffers R, Longerich T, Breuhahn K, et al. Posttranscriptional destabilization of the liver-specific long noncoding RNA HULC by the IGF2 mRNA-binding protein 1 (IGF2BP1) Hepatology. 2013;58:1703–1712. doi: 10.1002/hep.26537. [DOI] [PubMed] [Google Scholar]
  • 46.Panzitt K, Tschernatsch MM, Guelly C, Moustafa T, Stradner M, Strohmaier HM, Buck CR, Denk H, Schroeder R, Trauner M, et al. Characterization of HULC, a novel gene with striking up-regulation in hepatocellular carcinoma, as noncoding RNA. Gastroenterology. 2007;132:330–342. doi: 10.1053/j.gastro.2006.08.026. [DOI] [PubMed] [Google Scholar]
  • 47.Liu Y, Pan S, Liu L, Zhai X, Liu J, Wen J, Zhang Y, Chen J, Shen H, Hu Z. A genetic variant in long non-coding RNA HULC contributes to risk of HBV-related hepatocellular carcinoma in a Chinese population. PLoS One. 2012;7:e35145. doi: 10.1371/journal.pone.0035145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Xie H, Ma H, Zhou D. Plasma HULC as a promising novel biomarker for the detection of hepatocellular carcinoma. Biomed Res Int. 2013;2013:136106. doi: 10.1155/2013/136106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.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. doi: 10.1074/jbc.M112.410506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Shibata C, Otsuka M, Kishikawa T, Yoshikawa T, Ohno M, Takata A, Koike K. Current status of miRNA-targeting therapeutics and preclinical studies against gastroenterological carcinoma. Molecular and Cellular Therapies. 2013;1:5. doi: 10.1186/2052-8426-1-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez-Torres M, Patel K, van der Meer AJ, Patick AK, Chen A, Zhou Y, et al. Treatment of HCV infection by targeting microRNA. N Engl J Med. 2013;368:1685–1694. doi: 10.1056/NEJMoa1209026. [DOI] [PubMed] [Google Scholar]
  • 52.Xu Y, Liu L, Liu J, Zhang Y, Zhu J, Chen J, Liu S, Liu Z, Shi H, Shen H, et al. A potentially functional polymorphism in the promoter region of miR-34b/c is associated with an increased risk for primary hepatocellular carcinoma. Int J Cancer. 2011;128:412–417. doi: 10.1002/ijc.25342. [DOI] [PubMed] [Google Scholar]
  • 53.Bao L, Yan Y, Xu C, Ji W, Shen S, Xu G, Zeng Y, Sun B, Qian H, Chen L, et al. MicroRNA-21 suppresses PTEN and hSulf-1 expression and promotes hepatocellular carcinoma progression through AKT/ERK pathways. Cancer Lett. 2013;337:226–236. doi: 10.1016/j.canlet.2013.05.007. [DOI] [PubMed] [Google Scholar]
  • 54.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. doi: 10.1016/j.jhep.2010.09.023. [DOI] [PubMed] [Google Scholar]
  • 55.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. doi: 10.1158/1078-0432.CCR-09-0092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Callegari E, Elamin BK, Giannone F, Milazzo M, Altavilla G, Fornari F, Giacomelli L, D’Abundo L, Ferracin M, Bassi C, et al. Liver tumorigenicity promoted by microRNA-221 in a mouse transgenic model. Hepatology. 2012;56:1025–1033. doi: 10.1002/hep.25747. [DOI] [PubMed] [Google Scholar]
  • 57.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. doi: 10.1002/hep.25576. [DOI] [PubMed] [Google Scholar]
  • 58.Wang Z, Gerstein M, Snyder M. RNA-Seq: a revolutionary tool for transcriptomics. Nat Rev Genet. 2009;10:57–63. doi: 10.1038/nrg2484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.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. doi: 10.1073/pnas.0907904107. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from World Journal of Hepatology are provided here courtesy of Baishideng Publishing Group Inc

RESOURCES