eISSN: 2449-8238
ISSN: 2392-1099
Clinical and Experimental Hepatology
Current issue Archive Manuscripts accepted About the journal Editorial board Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
2/2022
vol. 8
 
Share:
Share:
Original paper

Circulating HOTAIR potentially predicts hepatocellular carcinoma in cirrhotic liver and prefigures the tumor stage

Assem El-Shendidi
1
,
Rasha Ghazala
2
,
Ehab Hassouna
1

1.
Department of Internal Medicine (Hepatobiliary Unit), Faculty of Medicine, Alexandria University, Egypt
2.
Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Egypt
Clin Exp HEPATOL 2022; 8, 2: 139-146
Online publish date: 2022/06/02
Article file
- Circulating.pdf  [0.13 MB]
Get citation
 
PlumX metrics:
 

Introduction

Liver cancer is the sixth most commonly diagnosed cancer and the third leading cause of cancer-related deaths worldwide with most cases detected at late stages [1, 2]. Hepatocellular carcinoma (HCC) represents about 75-85% of primary liver cancers, and mostly develops in fibrotic chronic liver diseases (CLD), usually in cirrhosis [3].

Serum α-fetoprotein (AFP) is a gold standard biomarker mostly used for screening of suspicious HCC lesions in patients with liver cirrhosis, and has been proven to have capability of prefiguring the prognosis [4]. However, studies showed that AFP has a low diagnostic sensitivity, particularly in diagnosis of early-stage HCC, and its level is raised in other cases of CLD [4, 5]. Despite the recent progress in HCC diagnosis and intervention over the last decade, the overall prognosis still remains unsatisfactory, and there is an urgent need to search for novel biomarkers with high efficacy for early detection of HCC and prediction of tumor progression [6]. Elucidating the molecular mechanisms of tumor growth would allow the development of effective diagnostics and therapeutics, and ultimately help patients with HCC to achieve a favorable prognosis [7]. Research studies demonstrated that non-coding RNA plays a role in regulating multiple biological processes in various cancers including HCC [8, 9].

Only 1.5% of the human genome is protein coding while the remaining ~98% is transcribed into noncoding RNA (ncRNA) that does not produce proteins [10]. Long ncRNA (lncRNA), which consists of > 200 nucleotides, promotes the proliferation, migration, invasion, autophagy and apoptosis of tumor cells by regulating downstream target gene expression and cancer-related signaling pathways [11, 12]. Some lncRNAs may act as “oncogenes” or “tumor suppressors” for cancers and are useful in tumor diagnosis and prognosis [13]. In patients with HCC, lncRNA dysregulation was highly associated with disease recurrence, metastasis and poor clinical outcomes [14, 15].

Homeobox (HOX) transcript antisense intergenic RNA (HOTAIR) is a 2.2-kb gene residing in the mammalian HOXC locus on chromosome 12q13.13. It has been considered to play critical roles in the biological properties of various tumors [16]. It was discovered as a gene repressor of HOXD genes, and demonstrated the important relationship between epigenetic regulation by lncRNA and cancer [17]. Specifically, HOTAIR recruits the polycomb repressive complex 2 (PRC2) occupancy and enhances the trimethylated histone 3 lysine 27 (H3K27me3) repressive marks [18]. It plays an important role in the development and progression of complicated diseases, including esophageal squamous cell carcinoma, colon cancer and breast cancer [19]. Hypoxia regulates HOTAIR to modulate cell proliferation, programmed death, invasion and migration [20].

Therefore, the present study was designed to investigate the clinical usefulness of serum HOTAIR as a potential biomarker for predicting HCC and prefiguring the tumor stage.

Material and methods

Study population

This is a cross-sectional study conducted at the Alexandria University Hospital and included 80 patients with definite newly diagnosed HCC on top of hepatitis C virus (HCV)-related liver cirrhosis, divided into 40 HCC patients with tumor stages C-D according to the Barcelona Clinic Liver Cancer (BCLC) classification (group IA) and 40 HCC patients with tumor stages 0/A-B according to the BCLC classification (group IB), and 40 cirrhotic patients without HCC (group II). Also, 20 healthy subjects with no evidence of liver disease were included as a control (group III). Exclusion criteria were non-HCC hepatic malignancy, extra-hepatic malignancy, other causes of liver disease, and previous surgical, locoregional or systemic therapies of HCC.

The study was conducted in accordance with the provisions of the World Medical Association Declaration of Helsinki. The research protocol was approved by the Ethics Committee of the Alexandria Faculty of Medicine (IRB. No. 00007555). Informed consent was obtained from all subjects included in the study.

Preliminary evaluation

The study participants were evaluated clinically as regards manifestations of liver cirrhosis and malignancy, with complete clinical examination and routine laboratory investigations including complete blood picture and liver test assay. Serum AFP levels were measured using enzyme-linked immunosorbent assay (ELISA) (Cortez Diagnostics Inc., Cat. No. 5101Z) [4]. Liver disease severity was determined on the basis of the Child-Turcotte-Pugh (CTP) classification. The diagnosis of HCC was confirmed by triphasic computed tomography (CT) and/or dynamic magnetic resonance imaging (MRI) of the liver showing contrast hyper-enhancement in the arterial phase “wash-in” and hypo-enhancement in the portal or delayed phase “wash-out”, with specific focus on the tumor characteristics (location, maximum diameter, number of nodules, tumor-in-vein thrombus and metastasis) [21]. The stage of HCC was determined according to the BCLC classification [22].

Measurement of serum HOTAIR level

For laboratory evaluation of circulating HOTAIR expression, 10 ml of venous blood was withdrawn from every patient and control subject. Each blood sample was allowed to clot, then centrifuged at 1200 × g for 10 minutes to separate the serum sample, which was kept frozen at –80°C until use. Total RNA isolation from serum samples was performed using the Qiagen miRNeasy Mini-Kit (Applied Biosystems Inc., Cat. No. 217004). The concentration and purity of RNA were measured using nanodrop then complementary DNA (cDNA) was synthesized using the High Capacity cDNA Reverse Transcription Kit (Applied Biosystems Inc., Cat. No. Archive) [11]. The thermal cycle was programmed at 10 minutes hold at temperature 25°C, 120 minutes hold at temperature 37°C, 5 minutes hold at temperature 85°C, then lowering the temperature to 4°C and stopping the run. Each reaction comprised approximately 10 µg of RNA extract, 2 µl of RT Buffer, 0.8 µl of dNTP, 1 µl of reverse transcriptase, 1 µl of RNase inhibitor, 2 µl of RT random primers, and then the total volume was completed to 20 µl using nuclease-free water.

Following reverse transcription, cDNA was stored at –20°C to be used in real-time quantitative polymerase chain reaction (RT-qPCR) experiments. Thermo Scientific Maxima SYBR Green qPCR Master Mix (2X) kit (Thermo Fisher Scientific Inc., Cat. No. K0251) was used to perform RT-qPCR. For amplification of the lncRNA HOTAIR gene, the following sequences of forward and reverse primers were used: forward primer (HOTAIR-F), 5’-GGTAGAAAAAGCAACCACGAAGC-3’ and reverse primer (HOTAIR-R), 5’-ACATAAACCTCTGTCTGTGAGTGCC-3’. For amplification of the glyceraldehyde 3-phosphate dehydrogenase (GAPDH) gene as an endogenous control, the following sequences of forward and reverse primers were used: forward primer (GAPDH-F), 5’-GAAGGTGAAGGTCGGAGTCAAC-3’ and reverse primer (GAPDH-R), 5’-CAGAGTTAAAAGCAGCCCTGGT-3’. Each reaction contained 12.5 µl of Maxima SYBR Green qPCR Master Mix (2X), 1 µl of forward primer (50 pmol), 1 µl of reverse primer (50 pmol), 0.1 µl of ROX solution, 7.4 µl of nuclease-free water and 3 µl of cDNA. Samples were assayed in duplicate. RT-qPCR was programmed as follows: an initial activation step at 95°C for 10 minutes, followed by 3-step cycling (40 cycles): denaturation step at 95°C for 15 seconds, annealing step for 30 seconds at 63°C for lncRNA HOTAIR gene and 65°C for GAPDH gene, and finally an extension step at 72°C for 30 seconds.

Melting curve analysis was performed to verify specificity and identity of PCR products. Melting curve analysis was built into the software of real-time cyclers. Generally, melting curve data were analyzed between 59°C and 95°C. When the temperature was gradually increased, a sharp decrease in SYBR Green fluorescence was observed as the product underwent denaturation. Specific products were distinguished from nonspecific products by the difference in their melting temperatures. To avoid misinterpretation, negative controls were included in all runs. A blank control without DNA was included in each PCR run as a negative control. In order to avoid mispriming or other errors that might happen during PCR, Hot-Start PCR was used. Results were analyzed using StepOne Software v2.2. The fold change between a sample and a normal control for lncRNA HOTAIR was calculated with the relative quantification method (RQ = 2–ΔΔCT).

Statistical analysis

Data were fed to the computer and analyzed using the software SPSS Statistics version 20.0. (SPSS, Armonk, NY: IBM Corporation). Qualitative data were described as number and percentage. Quantitative data were described as range and mean ± standard deviation. The Kolmogorov-Smirnov test and Shapiro-Wilk test were used to verify the normality of data distribution. The MannWhitney U-test was used to compare between the groups for non-normally distributed numerical variables. Student’s t-test was used to compare between the groups for normally distributed numerical variables. The receiver operating characteristic (ROC) curve was plotted to determine the optimal cut-off value of the tested variables with the highest sensitivity and specificity for prediction of the outcome. The area under the curve (AUC), the positive predictive value (PPV) and the negative predictive value (NPV) were calculated for this cut-off value. Statistical significance of the obtained results was judged at the p < 0.05 level. All calculated p values were two-tailed.

Results

Baseline clinical and biochemical data of patients and healthy subjects included in the study are shown in Table 1.

Table 1

Distribution of clinical and biochemical data of patients and healthy subjects included in the study

Baseline characteristicsGroup IA (n= 40)Group IB (n= 40)Group II (n= 40)Group III (n= 20)
Age (years)
Range43-6845-6942-6744-68
Mean ±SD54.0 ±7.7361.0 ±4.9556.0 ±7.5555.2 ±7.24
Sex, n(%)
Male28 (70)26 (65)28 (70)14 (70)
Female12 (30)14 (35)12 (30)6 (30)
Hemoglobin level (g/dl)
Range6.1-15.97.7-15.26.1-14.8-14.9
Mean ±SD11.2 ±2.411.66 ±1.5510.4 ±2.212.6 ±1.4
Platelet count (× 103/mm3)
Range95-24186-21690-319135-392
Mean ±SD112.9 ±52.9119.73 ±35.68136.1 ±82.2241.5 ±56.4
Leucocyte count (× 103/mm3)
Range3-181-92-132-10
Mean ±SD7.8 ±3.15.41 ±1.887.3 ±2.66.2 ±2.3
Aspartate aminotransferase (U/l)
Range37.3-111.032.0-69.528.0-46.024.3-38.0
Mean ±SD71.05 ±20.1448.54 ±11.3335.87 ±4.7831.42 ±3.96
Alanine aminotransferase (U/l)
Range34.5-82.512.0-135.024.3-84.815.5-31.8
Mean ±SD71.05 ±20.1448.54 ±11.3335.87 ±4.7822.90 ±5.12
Albumin (g/dl)
Range1-42-41-63-5
Mean ±SD2.6 ±0.73.0 ±0.63.1 ±1.14.1 ±0.5
Bilirubin (mg/dl)
Range1.3-5.00.7-3.60.38-2.50.8-1.1
Mean ±SD3.17 ±1.192.23 ±0.8571.24 ±0.590.94 ±0.12
International normalized ratio
Range1.2-1.81.2-1.51.0-1.581.0-1.1
Mean ±SD1.48 ±0.181.37 ±0.1011.28 ±0.181.05 ±0.05

Serum AFP level

The mean serum AFP level was significantly higher in groups IA, IB and II compared to group III (p < 0.001). The mean serum AFP level was also significantly higher in group IA than groups IB and II (p = 0.003 and p = 0.001 respectively), but showed no significant difference between group IB and group II (p = 0.096) (Table 2).

Table 2

Statistical comparison between the groups of patients included in the study and healthy subjects as regard α-fetoprotein (AFP) serum level (ng/ml)

AFP (ng/ml)HCCGroup IIGroup III
Group IA (n= 40)Group IB (n= 40)(n= 40)(n= 20)
Min.-Max.45-180012.5-1000.03.50-150.04.0-7.60
Mean ±SD356.4 ±288.7260.49 ±248.8440.66 ±36.964.76 ±0.90
Median350.050.019.35.4
U (pcontrol)14.0* (< 0.001*)0.0* (< 0.001*)16.50* (< 0.001*)
Significance between groupsU (p1) = 91.50* (0.003*), U (p2) = 72.50*(0.001*), U (p3) = 138.0 (0.096)

[i] AFP – α-fetoprotein, HCC – hepatocellular carcinoma, U – Mann-Whitney test, p – value of comparison, Pcontrol – p value for comparing between control and each other group, p1 – p value for comparing between late and early HCC cases, p2 – p value for comparing between late HCC and cirrhosis, p3 – p value for comparing between early HCC and cirrhosis, *statistical significance at p ≤ 0.05

Serum HOTAIR expression

The mean serum HOTAIR expression was significantly higher in groups IA, IB and II compared to group III (p < 0.001). The mean serum HOTAIR expression was significantly higher in group IA than group IB (p = 0.013), and also was significantly higher in groups IA and 1B than group II (p < 0.001 and p = 0.018 respectively) (Table 3).

Table 3

Statistical comparison between the groups of patients included in the study and healthy subjects as regard 2ΔΔCt HOTAIR serum level

2–ΔΔCtHOTAIRHCCGroup II (n= 40)Group III (n= 20)
Group IA (n= 40)Group IB (n= 40)
Min.-Max.2.13-82.471.38-51.871.42-15.760.17-2.05
Mean ±SD32.12 ±27.0214.59 ±13.184.13 ±3.021.12 ±0.65
Median23.1212.765.191.04
U (pcontrol)3.0* (< 0.001*)14.0* (< 0.001*)22.0* (< 0.001*)
Significance between groupsU (p1) = 108.0* (0.013*), U (p2) = 41.0* (< 0.001*), U (p3) = 113.0* (0.018*)

[i] HOTAIR – homeobox transcript antisense intergenic RNA, HCC – hepatocellular carcinoma, U – Mann-Whitney test, p – value of comparison; pcontrol – p value for comparing between control and each other group, p1 – p value for comparing between late and early HCC cases, p2 – p value for comparing between late HCC and cirrhosis, p3 – p value for comparing between early HCC and cirrhosis, *statistical significance at p ≤ 0.05

Performance of serum HOTAIR expression to discriminate between HCC patients of BCLC stages 0/A-B and HCC patients of BCLC stages C-D

Serum AFP at cut-off value > 50 ng/ml (AUC = 0.73, p = 0.003) showed sensitivity of 60% and specificity of 80% in discriminating HCC patients of group IB from HCC patients of group IA. Serum HOTAIR at cut-off value > 15.45 (AUC = 0.71, p = 0.013) showed sensitivity of 66% and specificity of 78% in discriminating HCC patients of group IB from HCC patients of group IA. When serum HOTAIR was combined with AFP, the discriminative sensitivity and specificity increased to 74% and 90% respectively (AUC = 0.85, p < 0.001) (Table 4 and Fig. 1).

Table 4

Performance of circulating HOTAIR solely and combined with AFP to discriminate between HCC patients of BCLC stages 0/A-B and HCC patients of BCLC stages C-D

AUCp95% CICut-offSensitivitySpecificityPPVNPV
AFP0.73*0.003*0.723-0.819> 5060.08080.068.0
2-ΔΔct HOTAIR0.71*0.013*0.564-0.896> 15.4566.078.072.268.2
AFP and 2-ΔΔctHOTAIR0.85*< 0.001*0.789-0.9674.090.087.5075.0

[i] AFP – α-fetoprotein, HOTAIR – homeobox transcript antisense intergenic RNA, AUC – area under the curve, p – probability value, CI – confidence interval, PPV – positive predictive value, NPV – negative predictive value, *statistical significance at p ≤ 0.05

Fig. 1

Performance of circulating HOTAIR solely and combined with AFP to discriminate between HCC patients of BCLC stages 0/A-B and HCC patients of BCLC stages C-D

/f/fulltexts/CEH/47176/CEH-8-47176-g001_min.jpg

Performance of serum HOTAIR expression to discriminate between HCC patients of BCLC stages 0/A-B and patients with non-tumorous liver cirrhosis

Serum AFP at cut-off value > 150 ng/ml (AUC = 0.882, p < 0.001) showed sensitivity of 65% and specificity of 100% in discriminating HCC patients of group IB from patients with non-tumorous liver cirrhosis. Serum HOTAIR at cut-off value > 9.42 (AUC = 0.823, p < 0.001) showed sensitivity of 67.5% and specificity of 93.3% in discriminating HCC patients of group IB from patients with non-tumorous liver cirrhosis. When serum HOTAIR was combined with AFP, the discriminative sensitivity and specificity increased to 80% and 98.3% respectively (AUC = 0.954, p < 0.001) (Table 5 and Fig. 2).

Fig. 2

Performance of circulating HOTAIR solely and combined with AFP to discriminate between HCC patients of BCLC stages 0/A-B and patients with non-tumorous liver cirrhosis

/f/fulltexts/CEH/47176/CEH-8-47176-g002_min.jpg
Table 5

Performance of circulating HOTAIR solely and combined with AFP to discriminate between HCC patients of BCLC stages 0/A-B and patients with non-tumorous liver cirrhosis

AUCp95% CICut-offSensitivitySpecificityPPVNPV
AFP0.882< 0.001*0.802-0.938> 15065.010010081.1
2-ΔΔCTHOTAIR0.823< 0.001*0.734-0.892> 9.4267.593.387.181.2
AFP and 2-ΔΔct HOTAIR0.954*< 0.001*0.892-0.98680.098.394.187.9

[i] AFP – α-fetoprotein, HOTAIR – homeobox transcript antisense intergenic RNA, AUC – area under the curve, p – probability value, CI – confidence interval, PPV – positive predictive value, NPV – negative predictive value, *statistical significance at p ≤ 0.05

Discussion

LncRNA HOTAIR has been increasingly recognized as a major player in diverse cellular functions, and found to interact by epigenetic mechanisms to affect the chromatin state and regulate target genes [11, 16]. However, the role of HOTAIR in liver fibrosis remains unknown. The current study found that serum HOTAIR expression was significantly higher in cirrhotic patients compared to the healthy subjects. Similarly, HOTAIR expression was significantly increased in human fibrotic livers and activated hepatic stellate cells (HSC) through regulation of different physiological and pathological processes [23-25]. Moreover, HOTAIR expression level was increased in cirrhotic liver tissues from hepatitis B virus-infected patients and co-localized with smooth muscle actin alpha 2 (ACTA2), indicating that HSC may be the primary source for HOTAIR in fibrotic liver [23]. A functional characterization demonstrated that HOTAIR overexpression promoted cell proliferation and activation with up-regulation of fibrosis-related genes and downstream mediators, such as ACTA2, collagen type 1 alpha 1 (COL1A1), and matrix metalloproteinases (MMP) [23, 24]. Furthermore, HOTAIR may act as an endogenous ‘sponge’ of certain micro-RNA (miR) molecules, such as miR-148b and miR-29b, which regulate the expression of various signaling pathways involved in HSC activation and proliferation [23, 25]. These results suggest that HOTAIR suppression may represent a possible therapeutic target in liver fibrosis.

HOTAIR has been extensively studied as an oncogene in numerous cancers including HCC [9, 16, 19]. The current study found that serum HOTAIR was clearly over-expressed in patients with HCC compared to cirrhotic patients and healthy subjects, and the increase of serum HOTAIR was compatible with the advancement of HCC stage. Similarly, HOTAIR was significantly over-expressed in HCC compared to the adjacent non-cancerous cirrhotic liver tissues, and HOTAIR depletion significantly inhibited proliferation and invasion of liver cancer cells in vivo and in vitro [26-28]. Those findings suggested that HCC proliferation may be affected by HOTAIR expression in the cells. Likewise, HOTAIR was over-expressed in HCC as compared with the adjacent non-tumorous cirrhotic liver tissues and was associated with tumor size [29]. Recently, a study showed significantly higher expression of HOTAIR in tumor tissues compared to adjacent tissues and in peripheral blood of HCC patients compared to healthy controls, with a positive correlation between its expression in the tumor tissue and peripheral blood [30]. Moreover, HOTAIR was up-regulated in non-cirrhotic CLD patients, cirrhotic patients and HCC patients, and the level of increase was consistent with the progression of the disease toward HCC, and this was associated with advanced stages and higher grades of the tumor [31]. Although HOTAIR did not discriminate between cirrhotic cases and non-cirrhotic CLD cases, it showed significant capability to discriminate between cases of cirrhosis and HCC compared with healthy controls [31]. That study suggested that HOTAIR could be considered as a predictor and prognostic biomarker and a new therapeutic target for HCV-induced cirrhosis and HCC [31]. Intriguingly, HOTAIR expression significantly predicted the development of HCC in HCV-infected patients treated with direct-acting antiviral therapy, and the higher values correlated with poorer disease outcomes [32]. That study suggested that pre-treatment HOTAIR expression level could serve as a risk assessment biomarker for HCC in HCV-infected patients and should be rigorously taken into consideration [32].

The current study demonstrated that HOTAIR may potentially differentiate early-stage HCC from late-stage HCC. Likewise, HCC patients with higher HOTAIR expression in their tumors have shown an increased risk of lymph node metastasis and recurrence after hepatectomy [27]. Also, HOTAIR expression increased in both HCC and HepG2 cells, promoted tumor progression and was associated with earlier relapse [28, 29, 33]. Those results highlighted the importance of HOTAIR in the evolvement and the relapse of HCC. Intriguingly, the expression level of HOTAIR in HCC was significantly associated with poor tumor differentiation and early recurrence, whereas no associations of HOTAIR expression with cirrhosis, serum AFP level, and TNM stage of the tumor were revealed [28]. Notably, a high HOTAIR expression level was found to be an independent prognostic factor for predicting HCC recurrence in transplanted patients and shorter recurrence-free survival [26]. Also, it was found that HCC patients with low HOTAIR expression in tumor tissues and/or peripheral blood had significantly longer overall and progression-free survival and a favorable response to therapy in patients with advanced HCC [30].

Various mechanisms have been suggested to understand the oncogenic functions of HOTAIR gene expression in regulating HCC progression [34, 35]. Up-regulation of HOTAIR promoted the proliferation, migration, and invasion of human HCC cells through inducing certain autophagy-related genes (ATG), e.g. ATG3 and ATG7, which are indispensable for autophagosome formation and activation [29, 33, 36]. Also, HOTAIR plays a critical role in the progression of HCC via inhibition of RNA binding motif protein 38 (RBM-38) [37]. Moreover, in vitro assays in the HCC cell line Bel7402 demonstrated that knockdown of HOTAIR reduced cell proliferation and was associated with reduction in the levels of vascular endothelial growth factor (VEGF) protein and MMP-9, which are important for cell motility, tumor progression and metastasis [27]. Furthermore, HOTAIR reduces the expression of Wnt inhibitory factor 1 (WIF-1), causing activation of Wnt/β-catenin signaling, which indicates that HOTAIR is important in the progression and recurrence of HCC, partly through the regulation of this molecular mechanism [28, 38]. Additionally, in vitro and in vivo experiments of HOTAIR knockdown induced apoptosis, inhibited cell proliferation and migration, impeded the epithelial-mesenchymal transition, and finally reduced the metastatic potential of HCC [39]. Epigenetically, HOTAIR may sponge miR-217-5p and suppress miR-122 expression via DNA methylation, leading to promotion of tumorigenicity and cell progression in HCC [40, 41].

Conclusions

In the current study, HCC was clearly accompanied by over-expression of circulating HOTAIR and the level of this expression was significantly associated with tumor stage and progression. These results suggest that the circulating HOTAIR test may be used solely, or preferably in combination with the serum AFP test, to aid in HCC detection and staging. Hence, HOTAIR is a valuable clinical biomarker for HCC diagnosis and prediction of the clinical outcome. Also, it may help monitoring the response to treatment and detection of tumor relapse. Targeting HOTAIR may be a novel therapeutic target for precision therapy in HCC.

In order to correct for the limitations in the present work, it may be recommended that the validity of circulating HOTAIR expression to diagnose HCC in cirrhotic liver and differentiate the tumor stage should be extensively studied in prospective longitudinal studies of a large population including patients with other etiologies of CLD. Moreover, evaluation of HOTAIR expression in HCC tissue and peri-tumor cirrhotic liver tissue paired with blood expression in patients with different HCC grades and tumor stages would certainly help to fully explore its potential contribution to liver cancer progression and prognosis. Furthermore, it is imperative to assess its clinical value as a biomarker to help monitoring the response of HCC to treatment and detection of the tumor recurrence after surgical, locoregional or systemic therapies. Further studies to search for any significant role of HOTAIR expression in the pathophysiology of different stages of liver disease should be conducted as well.

Acknowledgements

The authors would like to deeply thank the Liver Cancer Multidisciplinary Team at the Alexandria Faculty of Medicine for their great help and support.

Disclosure

The authors declare no conflict of interest.

References

1 

Akinyemiju T, Abera S, Ahmed M, et al. Global Burden of Disease Liver Cancer Collaboration. 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: 1683-1691.

2 

Sung H, Ferlay J, Siegel RL, et al. 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.

3 

Llovet JM, Zucman-Rossi J, Pikarsky E, et al. Hepatocellular carcinoma. Nat Rev Dis Primers 2016; 2: 16018.

4 

Zhou L, Liu J, Luo F. Serum tumor markers for detection of hepatocellular carcinoma. World J Gastroenterol 2006; 12: 1175-1181.

5 

Takayama T, Makuuchi M, Kojiro M, et al. Early hepatocellular carcinoma: pathology, imaging, and therapy. Ann Surg Oncol 2008; 15: 972-978.

6 

Nakashima O, Kojiro M. Recurrence of hepatocellular carcinoma: Multicentric occurrence or intrahepatic metastasis? A view point in terms of pathology. J Hepatobiliary Pancreat Surg 2001; 8: 404-409.

7 

Hao K, Luk JM, Lee NP, et al. Predicting prognosis in hepatocellular carcinoma after curative surgery with common clinicopathologic parameters. BMC Cancer 2009; 9: 389.

8 

Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nat Rev Genet 2009; 10: 155-159.

9 

Klingenberg M, Matsuda A, Diederichs S, et al. Non-coding RNA in hepatocellular carcinoma: Mechanisms, biomarkers and therapeutic targets. J Hepatol 2017; 67: 603-618.

10 

Palazzo AF, Lee ES. Non-coding RNA: what is functional and what is junk? Front Genet 2015; 6: 2.

11 

Huarte M, Rinn JL. Large non-coding RNAs: missing links in cancer? Hum Mol Genet 2010; 19 (R2): R152-161.

12 

Yuan D, Chen Y, Li X, et al. Long non-coding RNAs: potential biomarkers and targets for hepatocellular carcinoma therapy and diagnosis. Int J Biol Sci 2021; 17: 220-235.

13 

Xu C, Qi R, Ping Y, et al. Systemically identifying and prioritizing risk lncRNAs through integration of pan-cancer phenotype associations. Oncotarget 2017; 8: 12041-12051.

14 

He Y, Meng XM, Huang C, et al. Long noncoding RNAs: Novel insights into hepatocellular carcinoma. Cancer Lett 2014; 344: 20-27.

15 

Li C, Chen J, Zhang K, et al. Progress and prospects of long noncoding RNAs (lncRNAs) in hepatocellular carcinoma. Cell Physiol Biochem 2015; 36: 423-434.

16 

Tang Q, Hann SS. HOTAIR: an oncogenic long non-coding RNA in human cancer. Cell Physiol Biochem 2018; 47: 893-913.

17 

Qi P, Du X. The long non-coding RNAs, a new cancer diagnostic and therapeutic gold mine. Mod Pathol 2013; 26: 155-165.

18 

Xu C-Z, Jiang C, Wu Q, et al. A Feed-forward regulatory loop between HuR and the long noncoding RNA HOTAIR promotes head and neck squamous cell carcinoma progression and metastasis. Cell Physiol Biochem 2016; 40: 1039-1051.

19 

Hajjari M, Salavaty A. HOTAIR: an oncogenic long noncoding RNA in different cancers. Cancer Biol Med 2015; 12: 1-9.

20 

Rinn JL, Kertesz M, Wang JK, et al. Functional demarcation of active and silent chromatin domains in human HOX loci by non-coding RNAs. Cell 2007; 129: 1311-1323.

21 

Gale PR, Forner A, Llovet JM, et al. European Association for the Study of the Liver. EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma. J Hepatol 2018; 69: 182-236.

22 

Llovet JM, Brú C, Bruix J. Prognosis of hepatocellular carcinoma: the BCLC staging classification. Semin Liver Dis 1999; 19: 329-338.

23 

Bian EB, Wang YY, Yang Y, et al. Hotair facilitates hepatic stellate cells activation and fibrogenesis in the liver. Biochim Biophys Acta Mol Basis Dis 2017; 1863: 674-686.

24 

Hanson A, Wilhelmsen D, DiStefano JK. The role of long non-coding RNAs (lncRNAs) in the development and progression of fibrosis associated with nonalcoholic fatty liver disease (NAFLD). Noncoding RNA 2018; 4: 18.

25 

Yu F, Chen B, Dong P, et al. HOTAIR epigenetically modulates PTEN expression via microRNA-29b: a novel mechanism in regulation of liver fibrosis. Mol Ther 2017; 25: 205-217.

26 

Yang Z, Zhou L, Wu LM, et al. Overexpression of long non-coding RNA HOTAIR predicts tumor recurrence in hepatocellular carcinoma patients following liver transplantation. Ann Surg Oncol 2011; 18: 1243-1250.

27 

Geng YJ, Xie SL, Li Q, et al. Large intervening non-coding RNA HOTAIR is associated with hepatocellular carcinoma progression. J Int Med Res 2011; 39: 2119-2128.

28 

Gao JZ, Li J, Du JL, et al. Long non-coding RNA HOTAIR is a marker for hepatocellular carcinoma progression and tumor recurrence. Oncol Lett 2016; 11: 1791-1798.

29 

Yang L, Zhang X, Li H, et al. The long noncoding RNA HOTAIR activates autophagy by upregulating ATG3 and ATG7 in hepatocellular carcinoma. Mol Biosyst 2016; 12: 2605-2612.

30 

Han C, Yang Y, Guo L, et al. The expression of long non-coding RNA HOTAIR in advanced hepatocellular carcinoma and its prognostic correlation with sunitinib therapy. Arch Med Sci 2022; 18: 71-78.

31 

Roshdy F, Farag MMS, El-Ahwany E, et al. Long non-coding RNA HOTAIR and HOTTIP as potential biomarkers for hepatitis C virus genotype 4-induced hepatocellular carcinoma. Egypt J Med Hum Genet 2020; 21: 7.

32 

El-Khazragy N, Elshimy AA, Hassan SS, et al. Lnc-HOTAIR predicts hepatocellular carcinoma in chronic hepatitis C genotype 4 following direct-acting antivirals therapy. Mol Carcinog 2020; 59: 1382-1391.

33 

Su DN, Wu SP, Chen HT, et al. HOTAIR, a long non-coding RNA driver of malignancy whose expression is activated by FOXC1, negatively regulates miRNA-1 in hepatocellular carcinoma. Oncol Lett 2016; 12: 4061-4067.

34 

Niu ZS, Niu XJ, Wang WH. Long non-coding RNAs in hepatocellular carcinoma: Potential roles and clinical implications. World J Gastroenterol 2017; 23: 5860-5874.

35 

Ishibashi M, Kogo R, Shibata K, et al. Clinical significance of the expression of long non-coding RNA HOTAIR in primary hepatocellular carcinoma. Oncol Rep 2013; 29: 946-950.

36 

Lin R, Maeda S, Liu C, et al. A large noncoding RNA is a marker for murine hepatocellular carcinomas and a spectrum of human carcinomas. Oncogene 2007; 26: 851-858.

37 

Ding C, Cheng S, Yang Z, et al. Long non-coding RNA HOTAIR promotes cell migration and invasion via down-regulation of RNA binding motif protein 38 in hepatocellular carcinoma cells. Int J Mol Sci 2014; 15: 4060-4076.

38 

Qu B, Liu BR, Du YJ, et al. Wnt/β-catenin signaling pathway may regulate the expression of angiogenic growth factors in hepatocellular carcinoma. Oncol Lett 2014; 7: 1175-1178.

39 

Topel H, Bagirsakci E, Comez D, et al. LncRNA HOTAIR overexpression induced downregulation of c-Met signaling promotes hybrid epithelial/mesenchymal phenotype in hepatocellular carcinoma cells. Cell Commun Signal 2020; 18: 110.

40 

Gong X, Zhu Z. Long noncoding RNA HOTAIR contributes to progression in hepatocellular carcinoma by sponging miR-217-5p. Cancer Biother Radiopharm 2020; 35: 387-396.

41 

Cheng D, Deng J, Zhang B, et al. LncRNA HOTAIR epigenetically suppresses miR-122 expression in hepatocellular carcinoma via DNA methylation. EBioMedicine 2018; 36: 159-170.

Copyright: © 2022 Clinical and Experimental Hepatology. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
 
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.