Minireviews Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Cases. Jan 26, 2023; 11(3): 528-533
Published online Jan 26, 2023. doi: 10.12998/wjcc.v11.i3.528
Exosomal miRNA in early-stage hepatocellular carcinoma
Zhi-Qiang Wu, Yi-Xin Zhu, Yin-Chu Zhan, Department of Surgery, The Second People's Hosptal of Quzhou, Quzhou 324000, Zhejiang Province, China
Yun Jin, Department of Surgery, The Second Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou 310009, Zhejiang Province, China
ORCID number: Yun Jin (0000-0002-3073-4043); Yin-Chu Zhan (0000-0002-6017-1907).
Author contributions: Wu ZQ and Zhu YX designed and reviewed the manuscript; Wu ZQ, Zhu YX, and Jin Y wrote the paper; All authors read and approved the manuscript; Wu ZQ and Zhu YX contributed equally to this work.
Supported by the Quzhou Science and Technology Guided Project Planning, No. 2019133.
Conflict-of-interest statement: We declare that we have no conflict of interest related to this work.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (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: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Yin-Chu Zhan, MD, Director, Surgeon, Department of Surgery, The Second People's Hosptal of Quzhou, No. 338 Xin'an Avenue, Qujiang District, Quzhou 324000, Zhejiang Province, China. qzhospzyc@163.com
Received: October 28, 2022
Peer-review started: October 28, 2022
First decision: December 19, 2022
Revised: December 26, 2022
Accepted: January 5, 2023
Article in press: January 5, 2023
Published online: January 26, 2023

Abstract

The incidence and mortality of hepatic carcinoma (HCC) remain high, and early diagnosis of HCC is seen as a key approach in improving clinical outcomes. However, the sensitivity and specificity of current early screening methods for HCC are not satisfactory. In recent years, research around exosomal miRNA has gradually increased, and these molecules have emerged as attractive candidates for early diagnosis and treatment of HCC. This review summarizes the feasibility of using miRNAs in peripheral blood exosomes as early diagnostic tools for HCC.

Key Words: Hepatic carcinoma, Early diagnosis, Exosomal miRNA, Biomarker

Core Tip: The prognosis of hepatic carcinoma (HCC) is poor and surgical resection is the only potential radical cure. Early diagnosis of HCC is a key approach in improving clinical outcomes. However, the sensitivity and specificity of current early screening methods for HCC are not satisfactory. Exosomal miRNAs have become a candidate for early diagnosis and treatment of HCC. This review summarizes the feasibility of using miRNAs in peripheral blood exosomes as early diagnostic tools for HCC.



INTRODUCTION

Hepatocellular carcinoma (HCC) is one of the most common malignant tumors in the world, with the seventh highest incidence and second highest mortality among all malignant cancer types[1,2]. About 905000 patients are newly diagnosed with liver cancers each year around the world, with 75% of these being accounted for by HCC alone, and altogether causing about 830000 deaths[3]. Infection with hepatitis B virus (HBV) is the most common cause of HCC, and more than half of the world’s new cases of HCC were detected in China[4]. The high incidence and mortality of HCC pose a significant threat to the health of Chinese people and place a heavy burden on society[5].

It is considered that the best stage for treatment of HCC is Stage I, or subclinical liver cancer[6]. However, HCC symptoms in this period are not obvious, and most patients are not diagnosed until the middle or late stages. Therefore, development of more effective screening methods that allow earlier detection of HCC represents an important research focus. The five-year survival rate for patients with early-stage HCC is more than 70%, but this rate drops to less than 12.5% for patients with advanced HCC[7].

Tumor markers represent one approach for the early diagnosis of HCC, allowing earlier detection of primary tumors, recurrent tumors, and metastatic tumors than other methods. Yet, the accuracy of prediction and diagnosis achieved by tumor markers could still be improved, and such improvements would confer parallel improvements in prognosis and patient outcomes (Table 1)[8-12]. Alpha-fetoprotein (AFP), which has been widely used as a marker for HCC diagnosis and monitoring, is not highly sensitive to early HCC, and may also be increased in patients without HCC but with chronic viral hepatitis and cirrhosis[13]. As such, AFP can lead to misdiagnoses or unclear clinical interpretation, particularly if detected alone[14]. Despite the obvious limitations of using AFP as an early screening marker for HCC, its ubiquity has precluded its replacement by other clinical markers. At present, a variety of molecular markers are used in the diagnosis of HCC, but they all have the problems of low sensitivity and insufficient specificity. The sensitivity of AFP-L3, a glycoform of AFP, for the diagnosis of early HCC is less than 28%[15]. Another biomarker, protein induced by vitamin K absence II (PIVKA II), is also relatively insensitive for diagnosing early HCC[7]. Therefore, it is an urgent research direction to identify tumor markers with high sensitivity and specificity for the early diagnosis of HCC.

Table 1 Biomarkers for the diagnosis of hepatocellular carcinoma.
Biomarker
Ref.
Country
Sensitivity (%)
Specificity (%)
AFPTrevisani et al[8]Italy6090
DCPFeng et al[9]China 8391
AFP-L3Toyoda et al[10]Japan4185
GP73Marrero et al[11]United States6288
miR-21-5pGhosh et al[12]India7468
RELATIONSHIP BETWEEN RNA AND HCC

In recent years, non-coding RNA has attracted ever-increasing attention in the field of biological medicine. Our understanding of these RNA molecules has progressed rapidly, yielding a new understanding of cellular life and providing new opportunities for the diagnosis and treatment of various diseases. Among such non-coding RNA, the most extensively studied, to date, is microRNA (miRNA) - a class of small non-coding single-stranded RNA molecules containing 19-25 nucleotides. They are formed from double-stranded RNA precursors composed of 70-100 nucleotides in a hairpin structure. The sequences of miRNA are highly conserved across different species, suggesting that these very small molecules may play important roles in various cellular processes such as development, proliferation, differentiation, and apoptosis[16]. Significant differences in miRNA expression profiles have also been identified in some diseases[17,18], indicating that miRNA may be used as biomarkers for the diagnosis and prognosis of such diseases, including malignant tumors. Due to their structure, miRNAs can stably exist in the blood circulation, but the form of miRNAs in the circulation is not clear. Since RNA exposed to the blood is degraded within a short period of time, some researchers have pointed out that the stable existence of miRNAs in plasma must indicate the presence of protective macromolecules. Increasing evidence has shown that the main components of such protective macromolecular complexes are exosomes[19].

The correlation between miRNA and HCC was first proposed by Murakami, who analyzed the expression of microRNAs in tumor and adjacent tissues of HCC patients, as well as the liver tissues of hepatitis patients. It was found that miRNA-99a was positively correlated with the degree of pathological differentiation of HCC, while miRNA-20, miRNA-18 and pre-miRNA-18 were negatively correlated with the differentiation of HCC and positively correlated with the occurrence of HCC[20]. Since then, additional studies have confirmed close associations between miRNAs and HCC. These miRNAs can be roughly divided into two categories: "non-liver-specific miRNAs," such as miRNA-21, miRNA-221/222, and let-7, which are abnormally expressed in various tumors such as liver cancer, pancreatic cancer, and lung cancer[21-23], and "liver-specific miRNAs," which are only abnormally expressed in HCC. An example of a liver-specific miRNA is miRNA-122, which is up-regulated in HCC and suppresses the expression of the proto-oncogene c-myc through transcriptional activators[24]. Studies have shown that miRNA-122 is up-regulated in 70% of human liver cancer tissues and 100% of liver cancer cell models[25].

HCC is the result of multiple genetic mutations, which can occur in oncogenes or tumor suppressor genes, growth factors or their receptors, and myriad signaling pathways controlling cellular proliferation or behavior. Gene mutations also play a very important role in the progression of a tumor. When the expression of a gene or a class of molecules is silenced or enhanced, the possibility of tumorigenesis is present[26], and microRNA is gradually becoming the focus of this kind of research. miRNAs - small RNA molecules with very simple structures that regulate hundreds of mRNAs - play an unusual role in gene expression networks. Abnormally expressed miRNA may play a role similar to oncogenes such as myc or tumor suppressor genes such as p53, inducing or inhibiting liver tumorigenesis according to the specific cellular function of the target gene or genes regulated by that specific miRNA[27]. Meanwhile, and critically, miRNA can also influence the therapeutic effects of chemotherapy and intervene in the process of drug tolerance[28,29].

Regarding the use of miRNAs as molecular markers for tumors, miRNA-21 has gained attention as the first miRNA used in the clinic[30]. Many studies have confirmed that miRNA-21 is an oncogene, promoting liver tumor growth and metastasis by inhibiting the tumor suppressor genes PTEN and MAP2K3[31,32]. In 2012, Tomimaru et al[33] found that expression of miRNA-21 was increased in the plasma and tumor tissues of HCC patients, and that there was a correlation between them. Further investigation revealed that plasma miRNA-21 had clinical application value and could be used to diagnose HCC. Several additional studies have also shown that various miRNAs can be used for the diagnosis or prognosis of HCC patients. For example, serum miRNA-122 can also be used as a tumor marker for the diagnosis of HCC[34], while miRNA-125 and miRNA-233 can further be used for the early diagnosis of HCC patients who are HBV-positive[35]. miR-140 can also be used to determine the prognosis of HCC patients[36].

EXOSOMAL MIRNA IN HCC

The stability of miRNAs in plasma depends on exosomes. Exosomes are bilayer lipid membrane-coated vesicles with diameters of about 30-100 nm that can be released out of the cell and into the blood, urine, saliva, and other body fluids. It is generally believed that exosomes are composed of such lipid molecules as well as myriad amino acids and proteins, among which the common markers of exosomes have been identified as CD9, CD63, CD81, CD82, and others[37]. Nucleic acids in exosomes include mRNA, DNA, miRNA and other non-coding RNAs. Exosomes can carry these functional substances between cells and mediate communication between cells, thus regulating protein synthesis, cellular proliferation and differentiation, antiviral activity, and myriad other physiological and pathological activities.

Exosomal miRNAs have multiple potential functions in cell-to-cell communication. As such, they can be used to detect pathophysiological changes in the body, track changes in tumors, and aid in the diagnosis and prognosis of various diseases[38]. Down-regulation of exosomal miRNA expression has been shown to play a certain role in the mechanism of tumorigenesis[39]. Because exosomes and their contents (mainly miRNAs) can reflect the state of the cell they were released from – including whether that cell was of a tumorigenic or healthy state - exosomal miRNAs may hold a high value in the clinical diagnosis of tumors[40]. Detection of exosomes derived from tumor cells and their miRNA levels may become a novel biological tool with clinical potential and utility in people at high risk of cancer[41].

Protein and miRNA profiles in exosomes produced by HCC cells have been shown to be significantly different from those produced by normal cells[42,43]. Such exosomes can be ingested and internalized by other cells to deliver genes with certain functions[44]. Similar to the screening of serum tumor markers for liver cancer, exosomal miRNA can also be used as a valuable, non-invasive biomarker to distinguish the type and grade of liver inflammation, and then assist in the early diagnosis of liver cancer. Studies have also shown that circulating miRNAs may become biomarkers for HCC diagnosis due to the large number of miRNA variants in HCC cells[45]. Ghosh et al[12] identified a liver-specific exosomal miRNA, miR-21-5p, as an early circulating diagnostic marker for HCC with low AFP. The sensitivity, specificity, and accuracy of miR-21-5p differential diagnosis of HCC are 74%, 68%, and 71%, respectively.

CONCLUSION

In conclusion, although many miRNAs have been identified as tumor markers for early diagnosis of liver cancer in recent years, most of them have some defects and deficiencies, meaning that there is no clear consensus on which one or few miRNAs can improve the early diagnostics of HCC. Remaining challenges include a lack of further study on the specificity and sensitivity of target miRNAs in the diagnosis of liver cancer, a lack of robust clinical comparison between candidate miRNAs and the current tumor marker, AFP, and an expensive and cumbersome detection method for target miRNAs in HCC patients. As increased attention is placed upon exosomal miRNAs and their application at home and abroad, it is necessary and urgent to fully explore and realize the potential for exosomal miRNAs in the early diagnosis and treatment of liver cancer in clinical practice.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C, C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Luo L, China; Mohammed HA, Saudi Arabia; Wang ZX, China S-Editor: Liu JH L-Editor: A P-Editor: Liu JH

References
1.  Mohammed HA, Almahmoud SA, Arfeen M, Srivastava A, El-Readi MZ, Ragab EA, Shehata SM, Mohammed SAA, Mostafa EM, El-khawaga HA, Khan RA. Phytochemical profiling, molecular docking, and in vitro anti-hepatocellular carcinoid bioactivity of Suaeda vermiculata extracts. Arab J Chem. 2022;15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
2.  Mohammed HA, Khan RA. Anthocyanins: Traditional Uses, Structural and Functional Variations, Approaches to Increase Yields and Products' Quality, Hepatoprotection, Liver Longevity, and Commercial Products. Int J Mol Sci. 2022;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 19]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
3.  Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50630]  [Cited by in F6Publishing: 43253]  [Article Influence: 14417.7]  [Reference Citation Analysis (47)]
4.  Feng RM, Zong YN, Cao SM, Xu RH. Current cancer situation in China: good or bad news from the 2018 Global Cancer Statistics? Cancer Commun (Lond). 2019;39:22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 733]  [Cited by in F6Publishing: 1019]  [Article Influence: 203.8]  [Reference Citation Analysis (0)]
5.  El-Serag HB, Rudolph KL. Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology. 2007;132:2557-2576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3846]  [Cited by in F6Publishing: 4102]  [Article Influence: 241.3]  [Reference Citation Analysis (2)]
6.  Bruix J, Sherman M; American Association for the Study of Liver Diseases. Management of hepatocellular carcinoma: an update. Hepatology. 2011;53:1020-1022.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5972]  [Cited by in F6Publishing: 6340]  [Article Influence: 487.7]  [Reference Citation Analysis (1)]
7.  Wang W, Wei C. Advances in the early diagnosis of hepatocellular carcinoma. Genes Dis. 2020;7:308-319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 154]  [Article Influence: 38.5]  [Reference Citation Analysis (0)]
8.  Trevisani F, D'Intino PE, Morselli-Labate AM, Mazzella G, Accogli E, Caraceni P, Domenicali M, De Notariis S, Roda E, Bernardi M. Serum alpha-fetoprotein for diagnosis of hepatocellular carcinoma in patients with chronic liver disease: influence of HBsAg and anti-HCV status. J Hepatol. 2001;34(4):570-575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 512]  [Cited by in F6Publishing: 475]  [Article Influence: 20.7]  [Reference Citation Analysis (0)]
9.  Feng H, Li B, Li Z, Wei Q, Ren L. PIVKA-II serves as a potential biomarker that complements AFP for the diagnosis of hepatocellular carcinoma. BMC Cancer. 2021;21(1):401.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 49]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
10.  Toyoda H, Kumada T, Tada T, Kaneoka Y, Maeda A, Kanke F, Satomura S. Clinical utility of highly sensitive Lens culinaris agglutinin-reactive alpha-fetoprotein in hepatocellular carcinoma patients with alpha-fetoprotein < 20 ng/mL. Cancer Sci. 2011;102(5):1025-1031.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 88]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
11.  Marrero JA, Romano PR, Nikolaeva O, Steel L, Mehta A, Fimmel CJ, Comunale MA, D'Amelio A, Lok AS, Block TM. GP73, a resident Golgi glycoprotein, is a novel serum marker for hepatocellular carcinoma. J Hepatol. 2005;43(6):1007-1012.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 256]  [Cited by in F6Publishing: 277]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
12.  Ghosh S, Bhowmik S, Majumdar S, Goswami A, Chakraborty J, Gupta S, Aggarwal S, Ray S, Chatterjee R, Bhattacharyya S, Dutta M, Datta S, Chowdhury A, Dhali GK, Banerjee S. The exosome encapsulated microRNAs as circulating diagnostic marker for hepatocellular carcinoma with low alpha-fetoprotein. Int J Cancer. 2020;147:2934-2947.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 66]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
13.  European Association for the Study of the Liver; European Organisation for Research and Treatment of Cancer. EASL-EORTC clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol. 2012;56:908-943.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4059]  [Cited by in F6Publishing: 4345]  [Article Influence: 362.1]  [Reference Citation Analysis (2)]
14.  Motomura T, Shirabe K, Mano Y, Muto J, Toshima T, Umemoto Y, Fukuhara T, Uchiyama H, Ikegami T, Yoshizumi T, Soejima Y, Maehara Y. Neutrophil-lymphocyte ratio reflects hepatocellular carcinoma recurrence after liver transplantation via inflammatory microenvironment. J Hepatol. 2013;58:58-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 288]  [Cited by in F6Publishing: 330]  [Article Influence: 30.0]  [Reference Citation Analysis (0)]
15.  Marrero JA, Feng Z, Wang Y, Nguyen MH, Befeler AS, Roberts LR, Reddy KR, Harnois D, Llovet JM, Normolle D, Dalhgren J, Chia D, Lok AS, Wagner PD, Srivastava S, Schwartz M. Alpha-fetoprotein, des-gamma carboxyprothrombin, and lectin-bound alpha-fetoprotein in early hepatocellular carcinoma. Gastroenterology. 2009;137:110-118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 462]  [Cited by in F6Publishing: 502]  [Article Influence: 33.5]  [Reference Citation Analysis (0)]
16.  Whittaker S, Marais R, Zhu AX. The role of signaling pathways in the development and treatment of hepatocellular carcinoma. Oncogene. 2010;29:4989-5005.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 608]  [Cited by in F6Publishing: 646]  [Article Influence: 46.1]  [Reference Citation Analysis (0)]
17.  Lander ES. Cutting the Gordian helix--regulating genomic testing in the era of precision medicine. N Engl J Med. 2015;372:1185-1186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 47]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
18.  Baltimore D, Berg P, Botchan M, Carroll D, Charo RA, Church G, Corn JE, Daley GQ, Doudna JA, Fenner M, Greely HT, Jinek M, Martin GS, Penhoet E, Puck J, Sternberg SH, Weissman JS, Yamamoto KR. Biotechnology. A prudent path forward for genomic engineering and germline gene modification. Science. 2015;348:36-38.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 402]  [Cited by in F6Publishing: 305]  [Article Influence: 33.9]  [Reference Citation Analysis (0)]
19.  Gibbings DJ, Ciaudo C, Erhardt M, Voinnet O. Multivesicular bodies associate with components of miRNA effector complexes and modulate miRNA activity. Nat Cell Biol. 2009;11:1143-1149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 741]  [Cited by in F6Publishing: 762]  [Article Influence: 50.8]  [Reference Citation Analysis (0)]
20.  Murakami Y, Yasuda T, Saigo K, Urashima T, Toyoda H, Okanoue T, Shimotohno K. Comprehensive analysis of microRNA expression patterns in hepatocellular carcinoma and non-tumorous tissues. Oncogene. 2006;25:2537-2545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 876]  [Cited by in F6Publishing: 880]  [Article Influence: 48.9]  [Reference Citation Analysis (0)]
21.  Huang Y, Yang YB, Zhang XH, Yu XL, Wang ZB, Cheng XC. MicroRNA-21 gene and cancer. Med Oncol. 2013;30:376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 77]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
22.  Garofalo M, Quintavalle C, Romano G, Croce CM, Condorelli G. miR221/222 in cancer: their role in tumor progression and response to therapy. Curr Mol Med. 2012;12:27-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 263]  [Article Influence: 21.9]  [Reference Citation Analysis (0)]
23.  Wang X, Cao L, Wang Y, Wang X, Liu N, You Y. Regulation of let-7 and its target oncogenes (Review). Oncol Lett. 2012;3:955-960.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 148]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
24.  Wang B, Hsu SH, Wang X, Kutay H, Bid HK, Yu J, Ganju RK, Jacob ST, Yuneva M, Ghoshal K. Reciprocal regulation of microRNA-122 and c-Myc in hepatocellular cancer: role of E2F1 and transcription factor dimerization partner 2. Hepatology. 2014;59:555-566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 91]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
25.  Fornari F, Gramantieri L, Giovannini C, Veronese A, Ferracin M, Sabbioni S, Calin GA, Grazi GL, Croce CM, Tavolari S, Chieco P, Negrini M, Bolondi L. 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)]
26.  Shiraha H, Yamamoto K, Namba M. Human hepatocyte carcinogenesis (review). Int J Oncol. 2013;42:1133-1138.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 98]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
27.  Lujambio A, Lowe SW. The microcosmos of cancer. Nature. 2012;482:347-355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 802]  [Cited by in F6Publishing: 859]  [Article Influence: 71.6]  [Reference Citation Analysis (0)]
28.  Meng F, Henson R, Lang M, Wehbe H, Maheshwari S, Mendell JT, Jiang J, Schmittgen TD, Patel T. Involvement of human micro-RNA in growth and response to chemotherapy in human cholangiocarcinoma cell lines. Gastroenterology. 2006;130:2113-2129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 738]  [Cited by in F6Publishing: 770]  [Article Influence: 42.8]  [Reference Citation Analysis (0)]
29.  Xia L, Zhang D, Du R, Pan Y, Zhao L, Sun S, Hong L, Liu J, Fan D. miR-15b and miR-16 modulate multidrug resistance by targeting BCL2 in human gastric cancer cells. Int J Cancer. 2008;123:372-379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 531]  [Cited by in F6Publishing: 546]  [Article Influence: 34.1]  [Reference Citation Analysis (0)]
30.  Lawrie CH, Gal S, Dunlop HM, Pushkaran B, Liggins AP, Pulford K, Banham AH, Pezzella F, Boultwood J, Wainscoat JS, Hatton CS, Harris AL. Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. Br J Haematol. 2008;141:672-675.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1258]  [Cited by in F6Publishing: 1278]  [Article Influence: 79.9]  [Reference Citation Analysis (0)]
31.  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: 2117]  [Article Influence: 124.5]  [Reference Citation Analysis (0)]
32.  Xu G, Zhang Y, Wei J, Jia W, Ge Z, Zhang Z, Liu X. MicroRNA-21 promotes hepatocellular carcinoma HepG2 cell proliferation through repression of mitogen-activated protein kinase-kinase 3. BMC Cancer. 2013;13:469.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 75]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
33.  Tomimaru Y, Eguchi H, Nagano H, Wada H, Kobayashi S, Marubashi S, Tanemura M, Tomokuni A, Takemasa I, Umeshita K, Kanto T, Doki Y, Mori M. Circulating microRNA-21 as a novel biomarker for hepatocellular carcinoma. J Hepatol. 2012;56:167-175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 259]  [Cited by in F6Publishing: 271]  [Article Influence: 22.6]  [Reference Citation Analysis (0)]
34.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 212]  [Cited by in F6Publishing: 234]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
35.  Giray BG, Emekdas G, Tezcan S, Ulger M, Serin MS, Sezgin O, Altintas E, Tiftik EN. Profiles of serum microRNAs; miR-125b-5p and miR223-3p serve as novel biomarkers for HBV-positive hepatocellular carcinoma. Mol Biol Rep. 2014;41:4513-4519.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 97]  [Article Influence: 9.7]  [Reference Citation Analysis (1)]
36.  Yang H, Fang F, Chang R, Yang L. MicroRNA-140-5p suppresses tumor growth and metastasis by targeting transforming growth factor β receptor 1 and fibroblast growth factor 9 in hepatocellular carcinoma. Hepatology. 2013;58:205-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 217]  [Article Influence: 19.7]  [Reference Citation Analysis (0)]
37.  Kwizera EA, O'Connor R, Vinduska V, Williams M, Butch ER, Snyder SE, Chen X, Huang X. Molecular Detection and Analysis of Exosomes Using Surface-Enhanced Raman Scattering Gold Nanorods and a Miniaturized Device. Theranostics. 2018;8:2722-2738.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 140]  [Article Influence: 23.3]  [Reference Citation Analysis (0)]
38.  Eldh M, Olofsson Bagge R, Lässer C, Svanvik J, Sjöstrand M, Mattsson J, Lindnér P, Choi DS, Gho YS, Lötvall J. MicroRNA in exosomes isolated directly from the liver circulation in patients with metastatic uveal melanoma. BMC Cancer. 2014;14:962.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 75]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
39.  Kogure T, Yan IK, Lin WL, Patel T. Extracellular Vesicle-Mediated Transfer of a Novel Long Noncoding RNA TUC339: A Mechanism of Intercellular Signaling in Human Hepatocellular Cancer. Genes Cancer. 2013;4:261-272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 243]  [Article Influence: 22.1]  [Reference Citation Analysis (0)]
40.  Li Y, Zhang L, Liu F, Xiang G, Jiang D, Pu X. Identification of endogenous controls for analyzing serum exosomal miRNA in patients with hepatitis B or hepatocellular carcinoma. Dis Markers. 2015;2015:893594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 69]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
41.  Julich H, Willms A, Lukacs-Kornek V, Kornek M. Extracellular vesicle profiling and their use as potential disease specific biomarker. Front Immunol. 2014;5:413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 53]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
42.  Chen W, Mao Y, Liu C, Wu H, Chen S. Exosome in Hepatocellular Carcinoma: an update. J Cancer. 2021;12:2526-2536.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
43.  Liu C, Wu H, Mao Y, Chen W, Chen S. Exosomal microRNAs in hepatocellular carcinoma. Cancer Cell Int. 2021;21:254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
44.  Wang F, Li L, Piontek K, Sakaguchi M, Selaru FM. Exosome miR-335 as a novel therapeutic strategy in hepatocellular carcinoma. Hepatology. 2018;67:940-954.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 185]  [Cited by in F6Publishing: 186]  [Article Influence: 31.0]  [Reference Citation Analysis (0)]
45.  Lang FM, Hossain A, Gumin J, Momin EN, Shimizu Y, Ledbetter D, Shahar T, Yamashita S, Parker Kerrigan B, Fueyo J, Sawaya R, Lang FF. Mesenchymal stem cells as natural biofactories for exosomes carrying miR-124a in the treatment of gliomas. Neuro Oncol. 2018;20:380-390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 158]  [Article Influence: 31.6]  [Reference Citation Analysis (0)]