Open Access

Different types of viral‑host junction found in HBV integration breakpoints in HBV‑infected patients

  • Authors:
    • Peng Ruan
    • Xiufang Dai
    • Jun Sun
    • Chunping He
    • Chao Huang
    • Rui Zhou
    • Zhuo Cao
    • Lan Ye
  • View Affiliations

  • Published online on: November 29, 2018     https://doi.org/10.3892/mmr.2018.9709
  • Pages: 1410-1416
  • Copyright: © Ruan et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study surveyed the characteristics of hepatitis B virus (HBV) integration in the liver genomes of patients with acute hepatitis B (AHB), carriers of inactive hepatitis B surface antigen (HBsAg), and patients with chronic hepatitis B (CHB) receiving antiviral treatment. ‘Short‑read’ whole genome sequencing (WGS) with an average of 4,879x coverage for HBV integration was performed in three patients with AHB, two carriers of inactive HBsAg, and 13 patients with CHB receiving antiviral treatment. Conventional polymerase chain reaction and Sanger sequencing were used to verify integration breakpoints supported by at least two paired‑end reads, and viral‑host chimeric transcripts were surveyed simultaneously. HBV integration breakpoints were 100% identified with an average of 138.2±379.9 breakpoints per sample. The numbers of HBV integration breakpoints were positively associated with the sequencing depth coverage numbers and levels of intrahepatic covalently closed circular DNA, respectively (P<0.0001 and P<0.0001). Four types of viral‑host junction in 14 HBV integration breakpoints were detected (two viral junctions mapped in the HBs gene, one in the Precore gene, and others within the HBx gene): Forward simple junction, reverse simple junction, forward and reverse complicated junction, and microhomology were found in many of the junctions. Expression of viral‑human chimeric transcripts was observed in several breakpoints, including the HBs gene. As a result, HBV can integrate into the host gene in the same manner as non‑homologous end joining and microhomology‑mediated end joining with numerous sites, and a close association may exist between HBV integration and patient prognosis. HBx integration may be indispensable for viral‑host chimeric transcription and HBsAg may be produced from integrated DNA.

Introduction

Hepatitis B virus (HBV) infection represents a major risk factor for the development of hepatocellular carcinoma (HCC), ranking fifth in global cancer incidence and representing the third leading cause of cancer-associated mortality (1). Following the infection of hepatoctyes, HBV relaxed-circle DNA (rcDNA) is transferred to the nucleus, where it forms covalently closed circular DNA (cccDNA). Within infected cells, the pregenomic RNA is then transcribed from the cccDNA and transported to the cytoplasm, where the mature capsids of the rcDNA are reverse transcribed and either secreted from the cells or returned to the nucleus to form the cccDNA pool. During this process, some HBV DNA genes integrate into the host chromosomal DNA. HBV integration is suspected to be one of the most important etiological events in HBV-induced HCC (2).

Previous isolation of HBV integration sites using polymerase chain reaction (PCR)-based methods, including Alu-PCR or ligation-mediated PCR, has suggested that the HBV insertional sites occur randomly throughout the genome, leading to the suggestion that there are no preferential integration sites with little oncogenic annotation (35). Through the application of whole genome sequencing (WGS), a large cohort of integration sites have been identified and, among them, a number of hotspots have been found, including TERT, MLL4, KMT2B, CCNE1, and FN1. This suggests that HBV may have preferential integration sites associated with distinct biological consequences, for example, altering the function of endogenous genes, causing genetic damage and chromosomal instability, which may lead to tumorigenesis in HBV-infected patients (69). In the present study, according to WGS detection in patients with HBV infection, several types of viral-host junction were we found in HBV integration breakpoints. Certain expression characteristics of viral-human chimeric transcripts may assist in further understanding the molecular mechanisms of HBV integration and its role in hepatocarcinogenesis.

Materials and methods

Patients and samples

Liver biopsy specimens were collected from a total of 18 patients from Renmin Hospital of Wuhan University (Wuhan, China) between March and December 2016, comprising 16 men and two women, aged between 21 and 43 (32.16±7.11) years old. The patients included three patients with acute hepatitis B (AHB) achieving virological seroclearance (VR) spontaneously (group 1), two inactive HBsAg carriers (group 2), 13 patients with chronic hepatitis B (CHB) receiving nucleos(t)ide analogs (adefovir, 10 mg/day or lamivudine, 100 mg/day) either as monotherapy or with pegylated-interferon (IFN)α (100 µg/week). Among the patients with CHB, six patients had primary treatment failure (group 3), five patients had achieved VR (group 4), and two patients had achieved both VR and HBsAg seroclearance and had not relapsed for >6 months (group 5) (10). Every enrolled patient signed an informed consent form approved by the Ethics Committee of Renmin Hospital of Wuhan University. The biopsy specimens were frozen in liquid nitrogen and then stored at −80°C until further experimental analysis.

HBV integration detection

Liver DNA was extracted from biopsy specimens using the QIAamp DNA Mini kit (Qiagen GmbH, Hilden, Germany). A ‘short-read’ WGS for HBV integration was performed by the Beijing Genomics Institute (Shenzhen, China), as previously reported (7). A cluster of multiple read pairs was considered to be a candidate HBV integration breakpoint when it was identified with close mapping positions, linking an end of the human genome and an end of the HBV genome.

PCR and Sanger sequencing validation

In order to confirm the newly identified events and detect the unknown HBV sequences within the chimeric fragments, conventional PCR and Sanger sequencing were used to verify the HBV integration breakpoints with reads N≥2 in the partial HBV integration breakpoints detected by WGS, the strategy of which is shown in Fig. 1. The PCR primers were designed based on WGS-assembled fragments, in which one primer was located in the human genome and the other in the HBV genome. The PCR mix was prepared as follows: 1 µl of DNA; 2 µl of 10X Taq buffer; 11.5 µl of H2O; 2.5 µl of dNTPs; 1 µl of forward and reverse primers (10 µM, respectively) and 1 µl of Taq™ enzyme (Takara Bio, Inc., Otsu, Japan). PCR was subjected to the following cycling conditions: Initial denaturation for 10 min at 95°C; 40 cycles of denaturation for 10 sec at 95°C, annealing for 10 sec and extension at 72°C, final extension for 7 min at 72°C. The forward and reverse primer sequences the denaturation temperature and the duration of extension were determined by preliminary tests. The PCR products were electrophoresed on a 1% agarose gel and were then extracted and sequenced by Sanger sequencing (Shanghai Sangon Biology Engineering Technology & Service Co., Ltd., Shanghai, China). Finally, the results of sequencing were compared with the HBV and human genomes using the Basic local Alignment Search Tool (BLAST; blast.ncbi.nlm.nih.gov/Blast.cgi).

Viral-host chimeric transcripts

Total RNA was extracted as previously described (11). Reverse transcription-PCR (RT-PCR) was used to synthesize cDNA according to the manufacturer's protocol (PrimeScript RT™ Reagent kit with gDNA Eraser, Takara Bio, Inc.). The expression of viral-human chimeric transcripts was surveyed by conventional PCR, the conditions of which were designed by the preliminary tests above, and Sanger sequencing. The glyceraldehyde-3-phosphate dehydrogenase (GAPDH) gene was used as a control. The primer sequence for GAPDH was GAPDH-A1, sense 5′-ACCACAGTCCATGCCATCAC-3′ and antisense 5′-TCCACCACCCTGTTGCTGTA-3′. The PCR amplification conditions of GAPDH consisted of initial denaturation at 95°C for 10 min, followed by 95°C for 10 sec, 60°C for 10 sec, and 72°C for 20 sec for 30 cycles. The PCR product was 452 bp.

Intrahepatic (IH) HBV covalently closed circular (cccDNA) quantification

The IH HBV cccDNA levels were measured using quantitative PCR analysis as described previously (12). The cccDNA copy number for the extracted liver samples is calculated by dividing the copies/µl. In order to illuminate the influence of variation in the amount of liver tissue among samples, β-globin, the housekeeping gene (LightCycler Control Kit DNA, Roche Diagnostics) used to calculate the number of cells based on one copy of β-globin per genome, was used to allow for the standardization of the extracted DNA and expression of HBV cccDNA as copies per cell (copies/cell) (12).

Statistical analysis

The statistical analysis was performed using SPSS 13.0 statistical software (SPSS, Inc., Chicago, IL, USA). Continuous variables are expressed as the mean ± standard deviation. Correlations were evaluated using Pearson's correlation test. P<0.05 (two-sided) was considered to indicate a statistically significant difference.

Results

IH cccDNA quantification

With a lower limit of 0.00024 copies/cell, IH cccDNA levels were detected in two patients with AHB, six patients with CHB with primary treatment failure, five patients achieving VR, and one patient achieving both VR and HBsAg seroclearance (38.41±106.18 copies/cell).

Identification of global HBV integrations in HBV-infected patients

The average sequencing depth coverage of WGS for HBV integration was 4,879×. HBV integration breakpoints were identified within all 18 patients with a total of 2,083 and with an average of 138.2±379.9 breakpoints per sample (1–1,596 breakpoints per sample), whereas the higher number was 248.5±57.3 in group 3 and 18.6±13.7 in group 4, respectively (Fig. 2). The number of HBV integration breakpoints was positively associated with the sequencing depth coverage and the IH cccDNA levels, respectively (P<0.0001 and P<0.0001; Fig. 3).

Characteristics of HBV integration breakpoints

A total of 14 putative insertions (Table I) with at least two supporting paired-end reads were selected for PCR analysis and successful validation of 100% of these integration sites was achieved (Fig. 4); the unknown HBV sequences (HBV2) were detected within the chimeric fragments of numbers 1, 2, 3, 4, 7, 8, 9, 10, 11, 13 and 14 (Fig. 5). The designed primer sequences of conventional PCR for these breakpoints and their detected characteristics are shown in Table II. There were four types of viral-host junction: Forward simple junction, found in the breakpoints of numbers 2, 3, 8 and 12; reverse simple junction, found in those of numbers 4, 5, 6, 9, 10, 11, 13 and 14; and forward and reverse complicated junctions, found in those of numbers 1 and 7, respectively (Fig. 5). Microhomology was found in several viral-cellular junctions. For example, 3 bp (GCT) of microhomology were found in breakpoint of number 1, 5 bp (AAAAG) of microhomology were found in number 2, 2 bp (GC) of microhomology were found in number 8, and 7 bp (GACCTTC) of microhomology were found in number 11. A detailed analysis of the inserted viral fragments revealed that, with the exception of the breakpoints of number 4 and number 7 mapped in the S gene and that of number six mapped in the Precore gene, the others were localized within the HBx gene. The 3′-end of HBx in several of these breakpoints was found to be deleted.

Table I.

Viral-host junctions of 14 HBV integration breakpoints with at least two supporting paired-end reads.

Table I.

Viral-host junctions of 14 HBV integration breakpoints with at least two supporting paired-end reads.

NumberCodeHBV (nt)GeneHostLocation (nt)Gene
1920961,782Xchr9100,940,295CORO2A|intron
2920961,820PreC/Xchr9101,030,026Intergenic
3739581,471P/Xchr3   2,002,381Intergenic
424454  689S/Pchr2216,281,034Intergenic
5328271,807Xchr16   51,320,015Intergenic
6328271,878PreCchr16   51,320,070Intergenic
7688922,360P/Schr10   34,836,578PARD3|intron
8688921,651Xchr14   19,828,316Intergenic
9688921,754Xchr14   57,417,625Intergenic
10688921,831Xchr14   57,417,636Intergenic
11688921,760Xchr16   34,990,970Intergenic
12942201,546X/Pchr9   98,928,137Intergenic
13956581,465X/PchrX   36,919,408Intergenic
14439851,513X/Pchr5   42,009,760Intergenic

[i] HBV, hepatitis B virus; chr, chromosome.

Table II.

Sequences of primers and reaction conditions of 14 HBV integration breakpoints with at least two supporting paired-end reads.

Table II.

Sequences of primers and reaction conditions of 14 HBV integration breakpoints with at least two supporting paired-end reads.

NumberPrimer sequenceDenaturation temperature (°C)Extension (sec)Amplification fragment length (bp)
1 5′-ACTTCGCTTCACCTCTGC-3′6414321
5′-TATGATGGCACCACTGCA-3′
2 5′-GAGGCGGTGTCTAGGAGA-3′5417415
5′-GGTCAAATTGTTTGGATAAA-3′
3 5′-GCAAAACTCATCGGGACT-3′5616360
5′-TTGTGATGACTTGCTGGA-3′
4 5′-AAGACCTGCACGATTC-3′5014310
5′-TATGCTCACTTCCACA-3′
5 5′-GTCTTGCCCAAGGTCTTA-3′5614305
5′-CAGATGGCGCACTAACAA-3′
6 5′-TGAGTAACTCCACAGAAGC-3′5610136
5′-CAAGAAATAGCCCCAACT-3′
7 5′-CCCGATACAGAGCAGAGG-3′6212270
5′-GGTCATGGCATGGGAAGA-3′
8 5′-CGCTTCTCCGCCTATTGT-3′5818435
5′-ACCCTGGCATCCCTGGTTC-3′
9 5′-ATGGCTGCTAGGCTGTGC-3′5825622
5′-CCATTCCCAACTTGAAGATTTA-3′
10 5′-AGTATAGCTTGCCTGAGT-3′5616354
5′-CTGCTCTGAGGCAATTAA-3′
11 5′-GCTTGGAGGCTTGAACAG-3′5612253
5′-AGAGCCCCTTGGAAAATA-3′
12 5′-GAGGTGGCAATGAGGTGAG-3′5610103
5′-CAGAGGTGAAGCGAAGTG-3′
13 5′-AAAACTCATCGGGACTGA-3′4815319
5′-CATTTTGTTGACCTGGAA-3′
14 5′-CTGTGCTGCCAACTGGAT-3′6012254
5′-GCTAAGTGGAGCTTATTTCA-3′
Expression of viral-human chimeric transcripts

Among the 14 viral-host junctions with at least two supporting paired-end reads, chimeric transcripts were observed in breakpoints of numbers 1, 3, 4, 5, 6, 8, 12 and 14 (Fig. 6).

Discussion

HBV, a notorious DNA virus using cccDNA as its stable transcriptional template for HBV mRNAs, can insert its genome into the human genome and induce multiple hepatocarcinogenesis events. Current antiviral drugs, including INF-α and tenofovir, cannot completely eliminate intrahepatic cccDNA, which has significant involvement in HBV infection relapse and the pathogenesis of HCC and cirrhosis (13). However, HBV integration, the process by which underlying cancer-driving genetic events, including somatic mutation, structural rearrangement and clonal expansion, and its in-depth mechanisms and timing remain to be fully elucidated.

HBV DNA integration events occur with low frequency (0.01–0.1% of infected hepatocytes) in transient infections (14). Compared with traditional PCR-based methods, which do not have the required sensitivity for the detection of virus-cell junctions with such a low frequency of occurrence, WGS may provide insights into HBV integration detection by yielding greater specificity and sensitivity (69). In the present study, using ‘short reads’ WGS with an average sequencing depth of 4,879×, it was found that the rates of HBV integration were all 100% in patients with AHB, carriers of inactive HBsAg, and patients with CHB achieving different prognoses following antiviral treatment. The average number of integration breakpoints was 138.2±379.9 per sample. There may be several reasons for detecting a greater number of integration breakpoints per sample in the present study compared with other studies (15). Firstly, a previous study indicated that the ability to identify HBV insertion events depended on the HBV insertion allele frequency and the sequencing depth and coverage (16). This was supported by the positive correlation between the number of HBV integration sites and the sequencing depth coverage in the present study. Secondly, it was not possible to acquire significant differences in the number of integration sites among different groups due to a small number of patients in each group. The positive association between the number of HBV integration sites and the IH cccDNA level, which is associated with the overall survival rate of patients (17), suggested that there exists a close association between the frequency of HBV integration and the prognosis of HBV-infected patients, which requires validation in further investigations with larger sample sizes.

During HBV infection, host chromosomal DNA double stranded breaks (DSBs), provoking cellular responses with the most deleterious effects, may be caused by oxidative DNA damage (18,19). Under this condition and in order to maintain DNA integrity, a mixture of DNA repair mechanisms for host chromosomes may be involved in HBV DNA integration, for example, homologous recombination repair (HRR), classical non-homologous end joining (c-NHEJ), alternative end joining [including microhomology-mediated end joining (MMEJ)] and single strand annealing (20). HRR is an evolutionarily conserved, error-free repair mechanism, using an undamaged sister chromatid as a template to accurately repair the damage. By contrast, c-NHEJ is an error-prone repair pathway, repairing DSBs by joining two non-homologous DNA segments together, which may lead to the potential risks of gene deletion, insertion, indirect or direct repeats, and the phenomenon that HBV DNA molecules integrate into the host chromosomal DNA. Enriched microhomology (MH) exists between human and HBV genome sequences, and MMEJ may be another important mechanism mediating virus integration processes (21).

In the present study, several forms of junction were found between viral and host genes: Forward and reverse simple junctions, and forward and reverse complicated junctions, suggesting that they may be formed by NHEJ, as NHEJ is typically associated with deletions and sometimes insertions of different sequences at the termini (22). Secondly, the finding of MH in viral-host junctions in the present study, for example, 3 bp (GCT) in number 1, 5 bp (AAAAG) in number 2, and 7 bp (GACCTTC) in number 11, indicates that MMEJ may be key in their formation (23). This finding is similar to the result of a previous study, where significant enrichment of MH sizes of 2 and 5 bp was found in the integration junction (24). Thirdly, several HBV integration breakpoints were observed within the HBx genome in the present study, confirming that the HBx gene had more integration opportunities than other regions of HBV, due to the existence of a large number of viral transcriptional regulators in the HBx gene (8).

In the viral-human chimeric transcription analysis, it was found that several transcribed viral-human sequences were located within the X gene, demonstrating that chimeric transcripts were observed only when the site of integration was at 3′-end of HBx and often when its deletion occurred (7). Several independent lines of evidence have demonstrated that HBsAg is not only expressed from the episomal cccDNA minichromosome, but also from transcripts arising from HBV DNA integrated into the host genome (25). In the present study, this was demonstrated by the expression of chimera transcription of number 4, where the breakpoint of the inserted viral fragment was mapped in the S gene. This finding was also in accordance with a previous report, in which serum HBsAg and IH cccDNA levels were not correlated in patients with HBeAg-negative CHB (26), whereas patients with HBeAg-negative CHB usually have a longer, mostly perinatal HBV infection history and are expected to have more extensive HBV DNA integration than HBeAg-positive cases (27).

There were several limitations in the present study. Firstly, the full HBV integration sequences of these sites were not detected, the left end of which may not exactly match the double-stranded linear DNA ends, but may instead include terminal truncations of ~100 bp (14,28). Secondly, in order to further validate the hypothesis that HBV integration sites may have another origin for HBsAg production, chimeric protein expression requires detection from the integrated S gene. Finally, further experiments with larger sample sizes are required to further elucidate the molecular mechanisms and tumorigenesis of HBV integration.

In conclusion, the present study showed that HBV integration was detected in 100% of HBV-infected patients with a high number of integration sites. A close association existed between HBV integration and the prognoses of patients. HBx integration may be indispensable for viral-host chimeric transcripts and the expression of the HBs-host chimeric transcript suggests that HBsAg may be produced from integrated DNA.

Acknowledgements

No applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

XFD and JS conducted the experiments. CPH conducted statistical analysis. CH and RZ designed all the primers and modified the English language of the manuscript. ZC and LY collected the patient data. PR wrote the manuscript and made substantial contributions to the design of the present study. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Liver tissue in the present study was collected from patients between March and December 2016 and written informed consent was provided. The present study was approved by the Ethics Committee of Renmin Hospital of Wuhan University in March 2016.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Forner A, Llovet JM and Bruix J: Hepatocellular carcinoma. Lancet. 379:1245–1255. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Lok AS and McMahon BJ: Chronic Hepatitis B. Hepatology. 45:507–539. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Murakami Y, Saigo K, Takashima H, Minami M, Okanoue T, Bréchot C and Paterlini-Bréchot P: Large scaled analysis of hepatitis B virus (HBV) DNA integration in HBV related hepatocellular carcinomas. Gut. 54:1162–1168. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Saigo K, Yoshida K, Ikeda R, Sakamoto Y, Murakami Y, Urashima T, Asano T, Kenmochi T and Inoue I: Integration of hepatitis B virus DNA into the myeloid/lymphoid or mixed-lineage leukemia (MLL4) gene and rearrangements of MLL4 in human hepatocellular carcinoma. Hum Mutat. 29:703–708. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Tamori A, Yamanishi Y, Kawashima S, Kanehisa M, Enomoto M, Tanaka H, Kubo S, Shiomi S and Nishiguchi S: Alteration of gene expression in human hepatocellular carcinoma with integrated hepatitis B virus DNA. Clin Cancer Res. 11:5821–5826. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Jiang Z, Jhunjhunwala S, Liu J, Haverty PM, Kennemer MI, Guan Y, Lee W, Carnevali P, Stinson J, Johnson S, et al: The effects of hepatitis B virus integration into the genomes of hepatocellular carcinoma patients. Genome Res. 22:593–601. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Sung WK, Zheng H, Li S, Chen R, Liu X, Li Y, Lee NP, Lee WH, Ariyaratne PN, Tennakoon C, et al: Genome-wide survey of recurrent HBV integration in hepatocellular carcinoma. Nat Genet. 44:765–769. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Toh ST, Jin Y, Liu L, Wang J, Babrzadeh F, Gharizadeh B, Ronaghi M, Toh HC, Chow PK, Chung AY, et al: Deep sequencing of the hepatitis B virus in hepatocellular carcinoma patients reveals enriched integration events, structural alterations and sequence variations. Carcinogenesis. 34:787–798. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Zhao LH, Liu X, Yan HX, Li WY, Zeng X, Yang Y, Zhao J, Liu SP, Zhuang XH, Lin C, et al: Genomic and oncogenic preference of HBV integration in hepatocellular carcinoma. Nat Commun. 7:129922016. View Article : Google Scholar : PubMed/NCBI

10 

Chinese Society of Hepatology and Chinese Society of Infectious Diseases. Chinese Medical Association, The guideline of prevention and treatment for chronic hepatitis B (2010 version). Zhonghua Gan Zang Bing Za Zhi (Chinese J Hepatol). 19:13–24. 2011.

11 

Belloni L, Allweiss L, Guerrieri F, Pediconi N, Volz T, Pollicino T, Petersen J, Raimondo G, Dandri M and Levrero M: IFN-α inhibits HBV transcription and replication in cell culture and in humanized mice by targeting the epigenetic regulation of the nuclear cccDNA minichromosome. J Clin Invest. 122:529–537. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Werle-Lapostolle B, Bowden S, Locarnini S, Wursthorn K, Petersen J, Lau G, Trepo C, Marcellin P, Goodman Z, Delaney WE 4th, et al: Persistence of cccDNA during the natural history of chronic hepatitis B and decline during adefovir dipivoxil therapy. Gastroenterology. 126:1750–1758. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Zheng Q, Bai L, Zheng S, Liu M, Zhang J, Wang T, Xu Z, Chen Y, Li J and Duan Z: Efficient inhibition of duck hepatitis B virus DNA by the CRISPR/Cas9 system. Mol Med Rep. 16:7199–7204. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Tu T, Budzinska MA, Vondran FWR, Shackel NA and Urban S: Hepatitis B virus DNA integration occurs early in the viral life cycle in an in vitro infection model via NTCP-dependent uptake of enveloped virus particles. J Virol. Feb 7–2018.(Epub ahead of print). View Article : Google Scholar

15 

Murakami Y, Minami M, Daimon Y and Okanoue T: Hepatitis B virus DNA in liver, serum, and peripheral blood mononuclear cells after the clearance of serum hepatitis B virus surface antigen. J Med Virol. 72:203–214. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Sims D, Sudbery I, Ilott NE, Heger A and Ponting CP: Sequencing depth and coverage: Key considerations in genomic analyses. Nat Rev Genet. 15:121–132. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Hosaka T, Suzuki F, Kobayashi M, Hirakawa M, Kawamura Y, Yatsuji H, Sezaki H, Akuta N, Suzuki Y, Saitoh S, et al: HBcrAg is a predictor of post-treatment recurrence of hepatocellular carcinoma during antiviral therapy. Liver Int. 30:1461–1470. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Dandri M, Burda MR, Bürkle A, Zuckerman DM, Will H, Rogler CE, Greten H and Petersen J: Increase in de novo HBV DNA integrations in response to oxidative DNA damage or inhibition of poly(ADP-ribosyl)ation. Hepatology. 35:217–223. 2002. View Article : Google Scholar : PubMed/NCBI

19 

Bill CA and Summers J: Genomic DNA double-strand breaks are targets for hepadnaviral DNA integration. Proc Natl Acad Sci USA. 101:11135–11140. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Hu Z, Zhu D, Wang W, Li W, Jia W, Zeng X, Ding W, Yu L, Wang X and Wang L: Genome-wide profiling of HPV integration in cervical cancer identifies clustered genomic hot spots and a potential microhomology-mediated integration mechanism. Nat Genet. 47:158–163. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Mladenov E, Magin S, Soni A and Iliakis G: DNA double-strand-break repair in higher eukaryotes and its role in genomic instability and cancer: Cell cycle and proliferation-dependent regulation. Semin. Cancer Biol. 37–38. 51–64. 2016.

22 

Hu X, Lin J, Xie Q, Ren J, Chang Y, Wu W and Xia Y: DNA double-strand breaks, potential targets for HBV integration. J Huazhong Univ Sci Technolog Med Sci. 30:265–270. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Hastings PJ, Ira G and Lupski JR: A microhomology-mediated break-induced replication model for the origin of human copy number variation. PLoS Genet. 5:e10003272009. View Article : Google Scholar : PubMed/NCBI

24 

Yoo S, Wang W, Wang Q, Fiel MI, Lee E, Hiotis SP and Zhu J: A pilot systematic genomic comparison of recurrence risks of hepatitis B virus-associated hepatocellular carcinoma with low- and high-degree liver fibrosis. BMC Med. 150:2142017. View Article : Google Scholar

25 

Wooddell CI, Yuen MF, Chan HL, Gish RG, Locarnini SA, Chavez D, Ferrari C, Given BD, Hamilton J, Kanner SB, et al: RNAi-based treatment of chronically infected patients and chimpanzees reveals that integrated hepatitis B virus DNA is a source of HBsAg. Sci Transl Med. 9:eaan02412017. View Article : Google Scholar : PubMed/NCBI

26 

Ruan P, Zhou B, Dai X, Sun Z, Guo X, Huang J and Gong Z: Predictive value of intrahepatic hepatitis B virus covalently closed circular DNA and total DNA in patients with acute hepatitis B and patients with chronic hepatitis B receiving anti-viral treatment. Mol Med Rep. 9:1135–1141. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Thompson AJ, Nguyen T, Iser D, Ayres A, Jackson K, Littlejohn M, Slavin J, Bowden S, Gane EJ, Abbott W, et al: Serum hepatitis B surface antigen and hepatitis B e antigen titers: Disease phase influences correlation with viral load and intrahepatic hepatitis B virus markers. Hepatology. 51:1933–1944. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Mason WS, Low HC, Xu C, Aldrich CE, Scougall CA, Grosse A, Clouston A, Chavez D, Litwin S, Peri S, et al: Detection of clonally expanded hepatocytes in chimpanzees with chronic hepatitis B virus infection. J Virol. 83:8396–8408. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2019
Volume 19 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ruan P, Dai X, Sun J, He C, Huang C, Zhou R, Cao Z and Ye L: Different types of viral‑host junction found in HBV integration breakpoints in HBV‑infected patients. Mol Med Rep 19: 1410-1416, 2019
APA
Ruan, P., Dai, X., Sun, J., He, C., Huang, C., Zhou, R. ... Ye, L. (2019). Different types of viral‑host junction found in HBV integration breakpoints in HBV‑infected patients. Molecular Medicine Reports, 19, 1410-1416. https://doi.org/10.3892/mmr.2018.9709
MLA
Ruan, P., Dai, X., Sun, J., He, C., Huang, C., Zhou, R., Cao, Z., Ye, L."Different types of viral‑host junction found in HBV integration breakpoints in HBV‑infected patients". Molecular Medicine Reports 19.2 (2019): 1410-1416.
Chicago
Ruan, P., Dai, X., Sun, J., He, C., Huang, C., Zhou, R., Cao, Z., Ye, L."Different types of viral‑host junction found in HBV integration breakpoints in HBV‑infected patients". Molecular Medicine Reports 19, no. 2 (2019): 1410-1416. https://doi.org/10.3892/mmr.2018.9709