Circulating Non-Coding RNAs as Potential Diagnostic Biomarkers in Hepatocellular Carcinoma

Introduction

Hepatocellular carcinoma (HCC), one of the most common cancers worldwide, lists as a primary cause of cancer-related deaths globally.1 Currently, most patients with HCC grow into advanced stages after diagnosis, at which point the efficacy of radiotherapy and chemotherapy is limited and the time of surgical treatment was missed.2 Traditional imaging techniques including computed tomography (CT), positron emission computed tomography (PET) and magnetic resonance imaging (MRI) are the most widely used screening tools for HCC;3 however, they have limited sensitivity and are less efficient for detecting early HCC.4 Moreover, as the most commonly used clinical serum biomarker for detection of HCC, alpha-fetoprotein (AFP) also lacks the desired sensitivity and specificity for early diagnosis of HCC.5 It has been reported that the serum levels of AFP remain normal in up to 40% of the patients with HCC, particularly during the early stage.6 Therefore, there is an urgent need to develop clinical biomarkers for the detection of early-stage HCC.

Recently, there is extensive evidence suggesting that several circulating ncRNAs including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) in body fluids, such as urine, plasma, serum or saliva, could be potential non-invasive biomarkers for the diagnosis of cancers.7,8 These cell-free ncRNAs in body fluids are proposed to be leaked from cells following cell injury and death or be secreted by extracellular vesicles, such as exosomes.9,10 Due to the fact that ncRNAs have highly specific expression among different tissues or diseases, circulating ncRNAs are considered to be tumor-specific.11,12

A growing body of research has illuminated that ncRNAs play essential roles in the occurrence and development of HCC.13 Abnormal expression of circulating ncRNAs in HCC may provide potential diagnostic biomarkers.14 In this review, we will summarize the available knowledge on the diagnostic potential of circulating miRNAs, lncRNAs and circRNAs in HCC, aiming at providing new insights into the diagnosis of HCC.

Origins of Circulating ncRNAs It has been demonstrated that circulating ncRNAs are packaged into various membrane-bound vesicles including exosomes and microvesicles, and apoptotic bodies.15 Lipoproteins complexes such as high-density lipoprotein and RNA-binding proteins such argonaute 2 are known to be another sources of circulating ncRNAs.16 Advantages of ncRNAs as Potential Biomarkers ncRNAs exhibit high stability in the circulation. Circulating ncRNAs are stably maintained in diverse biological fluids including cerebrospinal fluid and peripheral serum/plasma due to their protection in exosomes, microvesicles, apoptotic bodies, and protein complexes.17 In terms of circRNAs, they have excellent stability owning to the covalently closed RNA circle without 5ʹ end caps or 3ʹ poly tails.18 Circulating ncRNAs can be quantified, even at low amounts, by quantitative reverse transcription polymerase chain reaction (qRT-PCR) with high sensitivity, specificity, and high dynamic range, a technique readily available in clinical laboratories.19 Additionally, the global profiles of ncRNAs can be obtained in a single experiment using qRT-PCR panels, next-generation sequencing or microarrays. Circulating ncRNAs are known to have tissue-specific expression patterns and profound relationships to development and diseases.20 Detection Methods of Circulating ncRNAs

It has been reported that different sources of liquid biopsy including whole blood, plasma, and serum can be used to quantify circulating ncRNAs. Different approaches including qRT-PCR, RNA-sequencing, and microarray, have been developed to study ncRNA expression.

The most commonly used method to detect the expression of specific ncRNAs is qRT-PCR. This method allows evaluation of the expression of a few specific molecules but do not permit the discovery of new ncRNAs or provide an overview of all ncRNAs.21 RNA-sequencing is a high-throughput sequencing method which can be used to sequence the entire transcriptome, leading to the identification of new ncRNAs.22 Microarray-based technique is another powerful high-throughput method extensively used for ncRNA profiling, because of their ability to screen large number of ncRNAs simultaneously in large variety of samples. However, microarrays use already known sequences, thus making them unsuitable for the discovery of novel ncRNAs.23,24 MiRNA Biomarkers

MiRNAs, a class of small non-coding RNAs with a length of 18–25 nucleotides, are endogenous and function as negative regulators of gene expression at the post-transcriptional level by promoting the degradation of messenger RNA (mRNA) or repressing its translation.25 It has been well demonstrated that miRNAs are secreted by cells through exosomes and extracellular vesicles, and secreted miRNAs remain stable in bodily fluids.26 Abnormal expression of miRNAs has been identified in various kinds of malignancies, which can function as oncogenes or tumor suppressors.27,28

Over the past several years, numerous studies have focused on exploring the possibility of circulating miRNAs as potential biomarkers for HCC diagnosis (Table 1). For example, it has been shown that the plasma miRNA-21 levels in the 126 patients with HCC were significantly increased than in patients with chronic hepatitis and healthy volunteers, respectively.29 ROC analysis showed that the area under the curve (AUC) of miRNA-21 in plasma was 0.773 with 61.1% sensitivity and 83.3% specificity in distinguishing patients with HCC from patients with chronic hepatitis; the AUC of miRNA-21 in plasma was 0.953 with 87.3% sensitivity and 92.0% specificity in distinguishing patients with HCC from patients with healthy volunteers.29 Recently, another study evaluated the diagnostic value of circulating levels of miRNA-9-3p in hepatitis C virus infection (HCV)-related HCC patients. The results showed that at a cutoff point of 1.01, miRNA-9-3p can discriminate between HCC patients and healthy volunteers with a sensitivity of 91.43% and a specificity of 87.50%.30 The performance of serum miRNA-224 as diagnostic biomarker for HCC at early stage has been assessed.31 The results of ROC analysis demonstrated that the AUC was 0.880 with 86.5% sensitivity and 76.7% specificity for serum miR-224 in discriminating early-stage HCC from all three controls (liver cirrhosis (LC), chronic hepatitis B (CHB) and healthy subjects), higher than that for AFP.31

Table 1 Circulating miRNAs Serve as Potential Diagnostic Biomarkers for HCC

Combining detection of different biomarkers has been reported to be a useful strategy due to the increased sensitivity and specificity of each biomarker. Consistently, several studies have evaluated the clinical value of miRNA panel for the early diagnosis of HCC. It has been shown that a miRNA panel (miR-122, miR-192, miR-21, miR-223, miR-26a, miR-27a and miR-801) can differentiate HCC from healthy (AUC 0.941), chronic hepatitis B (AUC 0.842), and cirrhosis (AUC 0.884), respectively.32 Similarly, another study has demonstrated that the unique 3-miRNA signature (miR-92a-3p, miR-107, and miR-3126-5p) combined with AFP can serve as a sensitive, specific, and noninvasive biomarker for the diagnosis of HCC, especially in the patients at early stages or with low AFP level.33 A large-scale, multicentre, retrospective longitudinal study has been conducted to assess the performance of serum miRNA for HCC detection.34 The results revealed that a seven-miRNA classifier (miR-29a, miR-29c, miR-133a, miR-143, miR-145, miR-192, and miR-505) had significantly higher sensitivity than AFP to distinguish patients with HCC from healthy controls, inactive HBsAg carriers, chronic hepatitis B patients, and patients with hepatitis B-induced liver cirrhosis.34

LncRNA Biomarkers

LncRNAs, a diverse class of ncRNAs, are defined as untranslated RNA molecules greater than 200 nucleotides in length with low protein-coding potential.40 Mounting evidence have suggested that lncRNAs play important roles in regulating gene expression at transcriptional, posttranscriptional and epigenetic levels in the development and progression of tumors.41,42 More importantly, aberrant signatures of lncRNAs are developed as the potential diagnostic biomarkers and therapeutic targets of cancer due to their specific expression pattern in carcinomas.43,44

Recent studies have evaluated the possibility of circulating lncRNAs as diagnostic biomarkers for HCC (Table 2). Ma et al recruited a cohort of 146 participants including healthy volunteers (HVs) and patients with chronic hepatitis B (CHB), cirrhosis and HCC, and investigated the potential use of circulating lncRNA differentiation antagonizing non-protein coding RNA (DANCR) in plasma as a diagnostic biomarker for HCC. The results showed that plasma DANCR exhibited significantly increased discriminatory power for differentiating patients with HCC from HVs and non-HCC patients compared to AFP.45 Similarly, another study showed that LINC00978 was upregulated in tissues and serum of HCC patients.46 The results of ROC analysis indicated that the upregulation of LINC00978 had a high diagnostic value to distinguish HCC patients from patients with hepatitis and liver cirrhosis and healthy controls.46 In addition, Zeng et al investigated the clinical value and functional role of lncRNA DQ786243 (lncDQ) in the pathogenesis of HCC.47 It has been found that serum lncDQ level could differentiate HCC patients from healthy controls, with an AUC of 0.804.

Table 2 Circulating lncRNAs Serve as Potential Diagnostic Biomarkers for HCC

In terms of the combination of lncRNA and AFP, it has been reported that combination of lncRNA SPRY4 intronic transcript 1 (SPRY4-IT1) and AFP possessed a moderate ability for discrimination between HCC patients and controls.48 The AUC of the combined indicators (0.800) is higher than that of SPRY4-IT1 alone (0.702).48 More recently, it has been indicated that a combination of lncRNA ubiquitin conjugating enzyme E2 C pseudogene 3 (UBE2CP3) and AFP yielded an AUC of 0.933, which was improved as compared to lncRNA UBE2CP3 (0.839) or AFP (0.912) alone.49 Similarly, combination of lncRNA JPX and AFP possessed a promoted ability for discrimination between HCC patients and controls (AUC 0.905, 97.1% sensitivity, 72.2% specificity) compared to JPX alone (AUC 0.814, 100% sensitivity, 52.4% specificity).50

In terms of the panel of lncRNAs, Kamel et al demonstrated that the panel of serum lncRNA urothelial carcinoma associated-1 (lncRNA UCA1) and WD repeat containing, antisense to TP53 (WRAP53) is superior to the AFP in specificity for the diagnosis of patients with HCC.51 Moreover, a panel based on the expression of lncRNAs uc001ncr and AX800134 accurately diagnosed HBV-positive HCC from control.52 It is worth noting that the diagnostic performance of the panel remained high in patients with AFP ≤ 400 ng/mL or in patients with early HCC.52 Furthermore, a plasma panel of lncRNAs HULC and 00152 possessed a moderate ability to discrimination between HCC and control with an area under ROC value of 0.87.53

CircRNA Biomarkers

As a novel class of endogenous ncRNAs, circRNAs form covalently closed continuous loops without 3’ end poly (A) tails and 5’ end caps.62 Owing to the outstanding characteristics including high stability, abundant expression, tissue-specific expression pattern, and wide distribution in various body fluids, circRNAs have great potential as suitable biomarkers for disease diagnosis.63 Recently, several reports have shown the potential of circulating circRNAs as a clinical biomarker in HCC diagnosis (Table 3). It has been reported that the AUC for circ_104075 was 0.973 with a sensitivity of 96.0% and a specificity of 98.3% in distinguishing patients with HCC from patients with healthy controls.64 Moreover, plasma circSMARCA5 showed a high accuracy (AUC = 0.938, 0.853, 0.711) for diagnosing HCC from healthy controls, hepatitis and cirrhosis.65 It is worth noting that plasma circSMARCA5 presented a high accuracy (AUC = 0.847, 0.706) for detecting HCC with serum AFP below 200 ng/mL from those hepatitis and cirrhosis with AFP below 200 ng/mL.65 These results suggested that circSMARCA5 may serve as a potential diagnostic biomarker for HCC, especially in HCC patients with AFP blow the cutoff value.

Table 3 Circulating circRNAs Serve as Potential Diagnostic Biomarkers for HCC

Exosomes, a specific subtype of extracellular vesicles of 30–150 nm, secreted by almost all types of cells and widely distributed in body fluids, including blood.66 As an important carrier of cell-to-cell signal transmission, exosomes carry a variety of important signal molecules such as proteins, nucleic acids and lipids, and are closely related to the development and occurrence of many disease processes, especially in cancer.67 Recent studies have indicated that circRNAs are enriched and stable in exosomes and exosomal circRNAs show great potential as biomarkers in human diseases.68 ROC curve analysis revealed that serum exosomal circWHSC1,69 circ_0072088,70 and circANTXR171 exhibited a potential diagnostic value for discriminating patients with HCC from healthy controls, respectively. Moreover, several studies have focused on distinguishing patients with HCC from patients with benign liver diseases by exosomal circRNAs. It has been demonstrated that the AUC for serum exosomal circ_0028861 was 0.79 for discriminating HCC from chronic HBV and cirrhosis individuals.72 Similarly, plasma exosomal circ_0070396 could discriminate HCC individuals from patients with chronic hepatitis B and liver cirrhosis.73

The ability of a combined panel of circRNAs to discriminate HCC patients has also been evaluated. It has been identified that a plasma circRNA panel (circPanel) containing three circRNAs (circ_0000976, circ_0007750 and circ_0139897) could detect HCC patients in three sets from three hospitals in China.74 Specifically, circPanel showed a higher accuracy than AFP to distinguish individuals with HCC from controls with AUCs of 0.843–0.864.74 Notably, circPanel also performed well in detecting small-HCC (solitary, ≤3 cm), AFP-negative HCC and AFP-negative Small-HCC.74 Another study has investigated the differentially expressed circRNAs in human blood exosomes from patients with HCC and investigated their diagnostic value.75 The results showed that the AUCs of serum exosomal circ_0004001, circ_0004123, and circ_0075792 were 0.79, 0.73, and 0.76 with adequate sensitivity and specificity in discriminating HCC patients from the healthy controls.75 Combination of these circRNAs considerably improved the AUC value (0.89), sensitivity and specificity, suggesting that the three circRNA signatures could be utilized as a diagnostic biomarker in HCC.75

Future Directions Although aberrant expression profiles of circulating ncRNAs in patients with HCC have been well demonstrated in numerous studies, the sample size is relative small and further confirmation with large-sample, multicentre, prospective clinical trial is needed in the future. Current studies on the abnormal expressions of circulating ncRNAs in patients with HCC are all cross-sectional, the dynamic change of circulating ncRNAs levels during the progression of the disease has not been reported. Although accumulating evidence has detailed the close relationship between HCC and ncRNAs, little is known about the causal relationship between them. There are no universally accepted methods and assays for measuring the levels of circulating ncRNAs, which might result in the lack of consistency in various studies. Therefore, the methodology of the detection of circulating ncRNAs levels needs to be standardized for future clinical use. Conclusions

With the development of high-throughput technologies, a large number of biologically functional ncRNAs have now been identified. Accumulating evidence has indicated that ncRNAs can be used as novel biomarkers for the diagnosis and screening of diseases due to the molecular characteristics. Although the above described circulating ncRNAs have adequate sensitivity and specificity in discriminating patients with HCC, it is necessary to cross-check and validate these markers with various cohorts of patients representing high variabilities of HCCs, including patients with different etiologies, HCC cellular profiles, and stages of cancer.

Acknowledgments

The present study was supported by the Shanghai Municipal Health Commission (No. 202040180 and 202040187).

Disclosure

The author declares that there are no conflicts of interest in this work.

References

1. Akinyemiju T, Abera S, Ahmed M, et al. The burden of primary liver cancer and underlying etiologies from 1990 to 2015 at the global, regional, and national level: results from the global burden of disease study 2015. JAMA Oncol. 2017;3(12):1683–1691. doi:10.1001/jamaoncol.2017.3055

2. Yan T, Yu L, Zhang N, et al. The advanced development of molecular targeted therapy for hepatocellular carcinoma. Cancer Biol Med. 2022;19:1–16. doi:10.20892/j.issn.2095-3941.2021.0661

3. Jiang H, Chen J, Xia C, Cao L, Duan T, Song B. Noninvasive imaging of hepatocellular carcinoma: from diagnosis to prognosis. World J Gastroenterol. 2018;24(22):2348–2362. doi:10.3748/wjg.v24.i22.2348

4. Lo E, Rucker A, Federle M. Hepatocellular carcinoma and intrahepatic cholangiocarcinoma: imaging for diagnosis, tumor response to treatment and liver response to radiation. Semin Radiat Oncol. 2018;28(4):267–276. doi:10.1016/j.semradonc.2018.06.010

5. Sun W, Liu Y, Shou D, et al. AFP (alpha fetoprotein): who are you in gastrology? Cancer Lett. 2015;357(1):43–46. doi:10.1016/j.canlet.2014.11.018

6. Daniele B, Bencivenga A, Megna A, Tinessa V. Alpha-fetoprotein and ultrasonography screening for hepatocellular carcinoma. Gastroenterology. 2004;127:S108–112. doi:10.1053/j.gastro.2004.09.023

7. Anfossi S, Babayan A, Pantel K, Calin G. Clinical utility of circulating non-coding RNAs - an update. Nat Rev Clin Oncol. 2018;15(9):541–563. doi:10.1038/s41571-018-0035-x

8. Grimaldi A, Zarone M, Irace C, et al. Non-coding RNAs as a new dawn in tumor diagnosis. Semin Cell Dev Biol. 2018;78:37–50. doi:10.1016/j.semcdb.2017.07.035

9. Southwood D, Singh S, Chatterton Z. Brain-derived cell-free DNA. Neural Regener Res. 2022;17(10):2213–2214. doi:10.4103/1673-5374.335794

10. Cinque A, Vago R, Trevisani F. Circulating RNA in kidney cancer: what we know and what we still suppose. Genes. 2021;12(6):835. doi:10.3390/genes12060835

11. Esteller M. Non-coding RNAs in human disease. Nat Rev Genet. 2011;12(12):861–874. doi:10.1038/nrg3074

12. Wang W, Han C, Sun Y, Chen T, Chen Y. Noncoding RNAs in cancer therapy resistance and targeted drug development. J Hematol Oncol. 2019;12(1):55. doi:10.1186/s13045-019-0748-z

13. Zhou H, Xu Q, Ni C, et al. Prospects of noncoding RNAs in hepatocellular carcinoma. Biomed Res Int. 2018;2018:6579436. doi:10.1155/2018/6579436

14. Song T, Li L, Wu S, et al. Peripheral blood genetic biomarkers for the early diagnosis of hepatocellular carcinoma. Front Oncol. 2021;11:583714. doi:10.3389/fonc.2021.583714

15. Wang J, Wang X, Song Y, et al. Circulating noncoding RNAs have a promising future acting as novel biomarkers for colorectal cancer. Dis Markers. 2019;2019:2587109. doi:10.1155/2019/2587109

16. Butz H. Circulating noncoding RNAs in pituitary neuroendocrine tumors-two sides of the same coin. Int J Mol Sci. 2022;23(9):5122. doi:10.3390/ijms23095122

17. Videira R, da Costa Martins P, Falcão-Pires I. Non-coding RNAs as blood-based biomarkers in cardiovascular disease. Int J Mol Sci. 2020;21(23):9285. doi:10.3390/ijms21239285

18. Xu Y, Chen F. Current status of functional studies on circular RNAs in rheumatoid arthritis and their potential role as diagnostic biomarkers. J Inflamm Res. 2021;14:1185–1193. doi:10.2147/JIR.S302846

19. Carballo-Perich L, Puigoriol-Illamola D, Bashir S, et al. Clinical parameters and epigenetic biomarkers of plaque vulnerability in patients with carotid stenosis. Int J Mol Sci. 2022;23(9):5149. doi:10.3390/ijms23095149

20. Kuo M, Liu S, Hsu Y, Wu R. The role of noncoding RNAs in Parkinson’s disease: biomarkers and associations with pathogenic pathways. J Biomed Sci. 2021;28(1):78. doi:10.1186/s12929-021-00775-x

21. Villa C, Stoccoro A. Epigenetic peripheral biomarkers for early diagnosis of Alzheimer’s disease. Genes. 2022;13(8):1308. doi:10.3390/genes13081308

22. Mortazavi A, Williams B, McCue K, Schaeffer L, Wold B. Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Methods. 2008;5(7):621–628. doi:10.1038/nmeth.1226

23. Schena M, Shalon D, Davis R, Brown P. Quantitative monitoring of gene expression patterns with a complementary DNA microarray. Science. 1995;270(5235):467–470. doi:10.1126/science.270.5235.467

24. Irfan J, Febrianto M, Sharma A, et al. DNA methylation and non-coding RNAs during tissue-injury associated pain. Int J Mol Sci. 2022;23(2):752. doi:10.3390/ijms23020752

25. Li Z, Rana T. Therapeutic targeting of microRNAs: current status and future challenges. Nat Rev Drug Discov. 2014;13(8):622–638. doi:10.1038/nrd4359

26. Mohr A, Mott J. Overview of microRNA biology. Semin Liver Dis. 2015;35(1):3–11. doi:10.1055/s-0034-1397344

27. Tang S, Li S, Liu T, et al. MicroRNAs: emerging oncogenic and tumor-suppressive regulators, biomarkers and therapeutic targets in lung cancer. Cancer Lett. 2021;502:71–83. doi:10.1016/j.canlet.2020.12.040

28. Babaei K, Shams S, Keymoradzadeh A, et al. An insight of microRNAs performance in carcinogenesis and tumorigenesis; an overview of cancer therapy. Life Sci. 2020;240:117077. doi:10.1016/j.lfs.2019.117077

29. Tomimaru Y, Eguchi H, Nagano H, et al. Circulating microRNA-21 as a novel biomarker for hepatocellular carcinoma. J Hepatol. 2012;56(1):167–175. doi:10.1016/j.jhep.2011.04.026

30. Wahb A, El Kassas M, Khamis A, Elhelbawy M, Elhelbawy N, Habieb M. Circulating microRNA 9-3p and serum endocan as potential biomarkers for hepatitis C virus-related hepatocellular carcinoma. World J Hepatol. 2021;13(11):1753–1765. doi:10.4254/wjh.v13.i11.1753

31. Lin L, Lu B, Yu J, Liu W, Zhou A. Serum miR-224 as a biomarker for detection of hepatocellular carcinoma at early stage. Clin Res Hepatol Gastroenterol. 2016;40(4):397–404. doi:10.1016/j.clinre.2015.11.005

32. Zhou J, Yu L, Gao X, et al. Plasma microRNA panel to diagnose hepatitis B virus-related hepatocellular carcinoma. J Clin Oncol. 2011;29(36):4781–4788. doi:10.1200/JCO.2011.38.2697

33. Zhang Y, Li T, Qiu Y, et al. Serum microRNA panel for early diagnosis of the onset of hepatocellular carcinoma. Medicine. 2017;96(2):e5642. doi:10.1097/MD.0000000000005642

34. Lin X, Chong Y, Guo Z, et al. A serum microRNA classifier for early detection of hepatocellular carcinoma: a multicentre, retrospective, longitudinal biomarker identification study with a nested case-control study. Lancet Oncol. 2015;16(7):804–815. doi:10.1016/S1470-2045(15)00048-0

35. Amr K, Elmawgoud Atia H, Elazeem Elbnhawy R, Ezzat W. Early diagnostic evaluation of miR-122 and miR-224 as biomarkers for hepatocellular carcinoma. Genes Dis. 2017;4(4):215–221. doi:10.1016/j.gendis.2017.10.003

36. Amr K, Ezzat W, Elhosary Y, Hegazy A, Fahim H, Kamel R. The potential role of miRNAs 21 and 199-a in early diagnosis of hepatocellular carcinoma. Gene. 2016;575(1):66–70. doi:10.1016/j.gene.2015.08.038

37. Chen S, Mao Y, Chen W, et al. Serum exosomal miR-34a as a potential biomarker for the diagnosis and prognostic of hepatocellular carcinoma. J Cancer. 2022;13(5):1410–1417. doi:10.7150/jca.57205

38. Yang C, Du P, Lu W. MiR-424 acts as a novel biomarker in the diagnosis of patients with hepatocellular carcinoma. Cancer Biother Radiopharm. 2021. doi:10.1089/cbr.2020.4141

39. Mohamed A, Omar A, El-Awady R, et al. MiR-155 and MiR-665 role as potential non-invasive biomarkers for hepatocellular carcinoma in Egyptian patients with chronic hepatitis C virus infection. J Transl Intern Med. 2020;8(1):32–40. doi:10.2478/jtim-2020-0006

40. Shaath H, Vishnubalaji R, Elango R, et al. Long non-coding RNA and RNA-binding protein interactions in cancer: experimental and machine learning approaches. Semin Cancer Biol. 2022. doi:10.1016/j.semcancer.2022.05.013

41. Yang J, Liu F, Wang Y, Qu L, Lin A. LncRNAs in tumor metabolic reprogramming and immune microenvironment remodeling. Cancer Lett. 2022;543:215798. doi:10.1016/j.canlet.2022.215798

42. Yao R, Wang Y, Chen L. Cellular functions of long noncoding RNAs. Nat Cell Biol. 2019;21(5):542–551. doi:10.1038/s41556-019-0311-8

43. Chi Y, Wang J, Wang J, Yu W, Yang J. Long non-coding RNA in the pathogenesis of cancers. Cells. 2019;8(9):1015. doi:10.3390/cells8091015

44. Nandwani A, Rathore S, Datta M. LncRNAs in cancer: regulatory and therapeutic implications. Cancer Lett. 2021;501:162–171. doi:10.1016/j.canlet.2020.11.048

45. Ma X, Wang X, Yang C, et al. DANCR acts as a diagnostic biomarker and promotes tumor growth and metastasis in hepatocellular carcinoma. Anticancer Res. 2016;36(12):6389–6398. doi:10.21873/anticanres.11236

46. Xu X, Gu J, Ding X, et al. LINC00978 promotes the progression of hepatocellular carcinoma by regulating EZH2-mediated silencing of p21 and E-cadherin expression. Cell Death Dis. 2019;10(10):752. doi:10.1038/s41419-019-1990-6

47. Zeng B, Lin Z, Ye H, et al. Upregulation of LncDQ is associated with poor prognosis and promotes tumor progression via epigenetic regulation of the EMT pathway in HCC. Cell Physiol Biochem. 2018;46(3):1122–1133. doi:10.1159/000488841

48. Jing W, Gao S, Zhu M, et al. Potential diagnostic value of lncRNA SPRY4-IT1 in hepatocellular carcinoma. Oncol Rep. 2016;36(2):1085–1092. doi:10.3892/or.2016.4859

49. Cao S, Huang J, Chen J, et al. Long non-coding RNA UBE2CP3 promotes tumor metastasis by inducing epithelial-mesenchymal transition in hepatocellular carcinoma. Oncotarget. 2017;8(39):65370–65385. doi:10.18632/oncotarget.18524

50. Ma W, Wang H, Jing W, et al. Downregulation of long non-coding RNAs JPX and XIST is associated with the prognosis of hepatocellular carcinoma. Clin Res Hepatol Gastroenterol. 2017;41(2):163–170. doi:10.1016/j.clinre.2016.09.002

51. Kamel M, Matboli M, Sallam M, Montasser I, Saad A, El-Tawdi A. Investigation of long noncoding RNAs expression profile as potential serum biomarkers in patients with hepatocellular carcinoma. Transl Res. 2016;168:134–145. doi:10.1016/j.trsl.2015.10.002

52. Wang K, Guo W, Li N, et al. Serum LncRNAs profiles serve as novel potential biomarkers for the diagnosis of HBV-positive hepatocellular carcinoma. PLoS One. 2015;10(12):e0144934. doi:10.1371/journal.pone.0144934

53. Li J, Wang X, Tang J, et al. HULC and Linc00152 act as novel biomarkers in predicting diagnosis of hepatocellular carcinoma. Cell Physiol Biochem. 2015;37(2):687–696. doi:10.1159/000430387

54. Kim S, Baek G, Ahn H, et al. Serum small extracellular vesicle-derived LINC00853 as a novel diagnostic marker for early hepatocellular carcinoma. Mol Oncol. 2020;14(10):2646–2659. doi:10.1002/1878-0261.12745

55. Tang J, Zhuo H, Zhang X, et al. A novel biomarker Linc00974 interacting with KRT19 promotes proliferation and metastasis in hepatocellular carcinoma. Cell Death Dis. 2014;5:e1549. doi:10.1038/cddis.2014.518

56. Wang X, Zhang W, Tang J, et al. LINC01225 promotes occurrence and metastasis of hepatocellular carcinoma in an epidermal growth factor receptor-dependent pathway. Cell Death Dis. 2016;7:e2130. doi:10.1038/cddis.2016.26

57. Lu J, Xie F, Geng L, Shen W, Sui C, Yang J. Investigation of serum lncRNA-uc003wbd and lncRNA-AF085935 expression profile in patients with hepatocellular carcinoma and HBV. Tumour Biol. 2015;36(5):3231–3236. doi:10.1007/s13277-014-2951-4

58. Huang R, Wang X, Zhang W, et al. Down-regulation of LncRNA DGCR5 correlates with poor prognosis in hepatocellular carcinoma. Cell Physiol Biochem. 2016;40:707–715. doi:10.1159/000452582

59. Yao Z, Jia C, Tai Y, et al. Serum exosomal long noncoding RNAs lnc-FAM72D-3 and lnc-EPC1-4 as diagnostic biomarkers for hepatocellular carcinoma. Aging. 2020;12(12):11843–11863. doi:10.18632/aging.103355

60. Konishi H, Ichikawa D, Yamamoto Y, et al. Plasma level of metastasis-associated lung adenocarcinoma transcript 1 is associated with liver damage and predicts development of hepatocellular carcinoma. Cancer Sci. 2016;107(2):149–154. doi:10.1111/cas.12854

61. Yu J, Han J, Zhang J, et al. The long noncoding RNAs PVT1 and uc002mbe.2 in sera provide a new supplementary method for hepatocellular carcinoma diagnosis. Medicine. 2016;95(31):e4436. doi:10.1097/MD.0000000000004436

62. Chen L, Yang L. Regulation of circRNA biogenesis. RNA Biol. 2015;12(4):381–388. doi:10.1080/15476286.2015.1020271

63. Zhang Z, Yang T, Xiao J. Circular RNAs: promising biomarkers for human diseases. EBioMedicine. 2018;34:267–274. doi:10.1016/j.ebiom.2018.07.036

64. Zhang X, Xu Y, Qian Z, et al. circRNA_104075 stimulates YAP-dependent tumorigenesis through the regulation of HNF4a and may serve as a diagnostic marker in hepatocellular carcinoma. Cell Death Dis. 2018;9(11):1091. doi:10.1038/s41419-018-1132-6

65. Li Z, Zhou Y, Yang G, et al. Using circular RNA SMARCA5 as a potential novel biomarker for hepatocellular carcinoma. Clinica chimica acta. 2019;492:37–44. doi:10.1016/j.cca.2019.02.001

66. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200(4):373–383. doi:10.1083/jcb.201211138

67. Milman N, Ginini L, Gil Z. Exosomes and their role in tumorigenesis and anticancer drug resistance. Drug Resist Updates. 2019;45:1–12. doi:10.1016/j.drup.2019.07.003

68. Tuo B, Chen Z, Dang Q, et al. Roles of exosomal circRNAs in tumour immunity and cancer progression. Cell Death Dis. 2022;13(6):539. doi:10.1038/s41419-022-04949-9

69. Lyu P, Zhai Z, Hao Z, Zhang H, He J. CircWHSC1 serves as an oncogene to promote hepatocellular carcinoma progression. Eur J Clin Invest. 2021;51(6):e13487. doi:10.1111/eci.13487

70. Lin Y, Zheng Z, Wang J, Zhao Z, Peng T. Tumor cell-derived exosomal Circ-0072088 suppresses migration and invasion of hepatic carcinoma cells through regulating MMP-16. Front Cell Dev Biol. 2021;9:726323. doi:10.3389/fcell.2021.726323

71. Huang C, Yu W, Wang Q, Huang T, Ding Y. CircANTXR1 contributes to the malignant progression of hepatocellular carcinoma by promoting proliferation and metastasis. J Hepatocell Carcinoma. 2021;8:1339–1353. doi:10.2147/JHC.S317256

72. Wang Y, Pei L, Yue Z, Jia M, Wang H, Cao L. The potential of serum exosomal hsa_circ_0028861 as the novel diagnostic biomarker of HBV-derived hepatocellular cancer. Front Genet. 2021;12:703205. doi:10.3389/fgene.2021.703205

73. Lyu L, Yang W, Yao J, et al. hsa_circ_0070396The diagnostic value of plasma exosomal for hepatocellular carcinoma. Biomark Med. 2021;15(5):359–371. doi:10.2217/bmm-2020-0476

74. Yu J, Ding W, Wang M, et al. Plasma circular RNA panel to diagnose hepatitis B virus-related hepatocellular carcinoma: a large-scale, multicenter study. Int J Cancer. 2020;146(6):1754–1763. doi:10.1002/ijc.32647

75. Sun X, Wang Y, Li G, Zhang N, Fan L. Serum-derived three-circRNA signature as a diagnostic biomarker for hepatocellular carcinoma. Cancer Cell Int. 2020;20:226. doi:10.1186/s12935-020-01302-y

76. Zhu C, Su Y, Liu L, Wang S, Liu Y, Wu J. Circular RNA hsa_circ_0004277 stimulates malignant phenotype of hepatocellular carcinoma and epithelial-mesenchymal transition of peripheral cells. Front Cell Dev Biol. 2020;8:585565. doi:10.3389/fcell.2020.585565

77. Chen W, Quan Y, Fan S, et al. Exosome-transmitted circular RNA hsa_circ_0051443 suppresses hepatocellular carcinoma progression. Cancer Lett. 2020;475:119–128. doi:10.1016/j.canlet.2020.01.022

78. Wang G, Liu W, Zou Y, et al. Three isoforms of exosomal circPTGR1 promote hepatocellular carcinoma metastasis via the miR449a-MET pathway. EBioMedicine. 2019;40:432–445. doi:10.1016/j.ebiom.2018.12.062

留言 (0)

沒有登入
gif