Integrated HBV DNA

整合 HBV DNA
  • 文章类型: Journal Article
    从乙型肝炎病毒(HBV)感染恢复后,再激活可以伴随免疫抑制而发生;因此,假设复制能力HBV在肝脏中持续存在。我们试图从13人自发恢复中检测持续性HBV。我们定量HBVDNA和RNA在核心肝活检(中位数1.72×106细胞)从注射药物(PWID)的人。在13个活检中,8(61%)有HBVDNA或RNA的证据和5(38%)有HBVDNA和RNA。来自cccDNA和整合的HBVDNA的mRNA。这里,我们显示流行的HBVDNA和RNA,尽管在PWID的临床恢复。
    我们使用一种灵敏的方法来确定从乙型肝炎病毒感染中恢复的13名个体的肝脏中乙型肝炎病毒DNA或RNA的量。尽管血液中没有检测到病毒,但仍有61%的人在肝脏中发现了病毒DNA或RNA。我们的发现支持从肝脏中消除所有乙型肝炎是一个困难的治疗目标。
    After recovery from a hepatitis B virus (HBV) infection, reactivation can occur with immunosuppression; thus, it is assumed that replication competent HBV persists in the liver. We sought to detect persistent HBV from 13 people with spontaneous recovery. We quantified HBV DNA and RNA in core liver biopsies (median 1.72x106 cells) from people who inject drugs (PWID). Among 13 biopsies, 8 (61%) had evidence of HBV DNA or RNA and 5 (38%) had both HBV DNA and RNA. mRNAs derived from cccDNA and integrated HBV DNA. Here, we show prevalent HBV DNA and RNA despite clinical recovery in PWID.
    We used a sensitive method to determine the amount of hepatitis B virus DNA or RNA in the livers of 13 individuals who recovered from hepatitis B virus infection. We found viral DNA or RNA in the liver in 61% of individuals despite no detectable virus in blood. Our findings support that eliminating all hepatitis B from the liver is a difficult treatment goal.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • Chronic hepatitis B virus (HBV) infection will greatly contribute to raising the occurrence probability of cirrhosis and hepatocellular carcinoma in patients. Although existing antiviral treatment regimens have a certain effect on delaying disease progression and improving prognosis, it is still not effective in attaining functional cures. Hepatitis B virus DNA integration may be one of the reasons for this phenomenon. Therefore, this paper reviews the possible mechanisms of HBV DNA integration in maintaining chronic inflammation of the liver, evading existing antiviral treatment methods, and inducing hepatocellular carcinoma so as to further deepen the understanding of the role of HBV DNA integration in the occurrence and development of chronic hepatitis B, providing ideas and references for formulating better treatment strategies.
    乙型肝炎病毒(HBV)慢性感染会导致患者发生肝硬化、肝细胞癌的概率大大增加。现有的抗病毒治疗方案虽然对延缓病情进展,改善预后有一定的作用,但仍不能有效地达到功能性治愈。整合HBV DNA可能是造成该现象的原因之一。因此,现就整合HBV DNA在维持肝脏慢性炎症,逃避现有的抗病毒治疗手段并且诱导肝细胞癌的可能作用机制进行综述,进一步加深对整合HBV DNA在慢性乙型肝炎发生和发展中作用的认识,为制定更优治疗策略提供思路和参考。.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    未经证实:HBV感染全球超过2.57亿人,并与肝细胞癌(HCC)的发展有关。HBVDNA整合到宿主基因组可能是肝癌发生的关键驱动因素。这里,我们利用靶向长读测序来确定HBVDNA整合的结构以及HBVmRNA的完整同工型信息,比传统的下一代测序平台更准确的定量。
    未经证实:从GS-US-174-0149临床试验中收集的新鲜冷冻肝活检中分离DNA和RNA。开发了生物素化寡核苷酸的泛基因型面板,以从剪切的基因组DNA(〜7kb)和来自聚腺苷酸化RNA的全长cDNA文库中富集HBV序列。在PacBio长读平台上对样品进行测序,并使用定制的生物信息学管道进行分析。
    UNASSIGNED:HBV靶向长读DNA测序产生了跨越整个整合的高覆盖率数据。引人注目的是,在42个样本中的13个(31%),我们能够检测到由2个不同染色体侧翼的HBV序列,表明与HBV整合相关的染色体易位。染色体易位对每个活检样本都是独特的,表明每个都是随机起源的,在某些情况下有克隆扩张的证据。使用靶向长读RNA测序,我们确定,在HBeAg阳性的患者中,超过95%的所有HBV转录本来自cccDNA。相比之下,HBeAg阴性的患者大多从整合中表达HBsAg。
    UNASSIGNED:靶向lso-Seq允许HBV转录组的准确定量和转录分配到cccDNA或整合起源。在非HCCCHB患者肝活检中存在多个独特的HBV相关染色体间易位,这表明具有诱变潜力的新机制可能有助于进展为HCC。
    UNASSIGNED:对HBV感染患者的新鲜冷冻肝活检进行靶向长读RNA和DNA测序。长读RNA测序捕获整个HBV转录本在一个单一的阅读,允许从HBV基因组重叠转录本的分辨率。该决议使我们能够量化来自整合的转录负担与cccDNA起源于个体患者。HBeAg阳性的患者与HBeAg阴性的患者相比,来自cccDNA的HBV转录组的比例明显更大。长读DNA测序捕获整个整合的HBV序列,包括单个读段内的数千碱基的侧翼宿主序列。该决议使我们能够描述两个不同宿主染色体两侧的整合事件,表明整合的HBVDNA与染色体间易位有关。这可能导致显著的转录失调和驱动进展为HCC。
    UNASSIGNED: HBV infects over 257 million people worldwide and is associated with the development of hepatocellular carcinoma (HCC). Integration of HBV DNA into the host genome is likely a key driver of HCC oncogenesis. Here, we utilise targeted long-read sequencing to determine the structure of HBV DNA integrations as well as full isoform information of HBV mRNA with more accurate quantification than traditional next generation sequencing platforms.
    UNASSIGNED: DNA and RNA were isolated from fresh frozen liver biopsies collected within the GS-US-174-0149 clinical trial. A pan-genotypic panel of biotinylated oligos was developed to enrich for HBV sequences from sheared genomic DNA (∼7 kb) and full-length cDNA libraries from poly-adenylated RNA. Samples were sequenced on the PacBio long-read platform and analysed using a custom bioinformatic pipeline.
    UNASSIGNED: HBV-targeted long-read DNA sequencing generated high coverage data spanning entire integrations. Strikingly, in 13 of 42 samples (31%) we were able to detect HBV sequences flanked by 2 different chromosomes, indicating a chromosomal translocation associated with HBV integration. Chromosomal translocations were unique to each biopsy sample, suggesting that each originated randomly, and in some cases had evidence of clonal expansion. Using targeted long-read RNA sequencing, we determined that upwards of 95% of all HBV transcripts in patients who are HBeAg-positive originate from cccDNA. In contrast, patients who are HBeAg-negative expressed mostly HBsAg from integrations.
    UNASSIGNED: Targeted lso-Seq allowed for accurate quantitation of the HBV transcriptome and assignment of transcripts to either cccDNA or integration origins. The existence of multiple unique HBV-associated inter-chromosomal translocations in non-HCC CHB patient liver biopsies suggests a novel mechanism with mutagenic potential that may contribute to progression to HCC.
    UNASSIGNED: Fresh frozen liver biopsies from patients infected with HBV were subjected to targeted long-read RNA and DNA sequencing. Long-read RNA sequencing captures entire HBV transcripts in a single read, allowing for resolution of overlapping transcripts from the HBV genome. This resolution allowed us to quantify the burden of transcription from integrations vs. cccDNA origin in individual patients. Patients who were HBeAg-positive had a significantly larger fraction of the HBV transcriptome originating from cccDNA compared with those who were HBeAg-negative. Long-read DNA sequencing captured entire integrated HBV sequences including multiple kilobases of flanking host sequence within single reads. This resolution allowed us to describe integration events flanked by 2 different host chromosomes, indicating that integrated HBV DNA are associated with inter-chromosomal translocations. This may lead to significant transcriptional dysregulation and drive progression to HCC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    在各种病毒感染中,通常观察到仅含有病毒糖蛋白的过量病毒颗粒(亚病毒颗粒或SVP)的产生,并且是免疫逃避的通常进化机制。在乙型肝炎病毒(HBV)感染,球形颗粒含有乙型肝炎表面抗原,数量超过传染性病毒10000-100000到1,对先天和适应性免疫反应有不同的抑制作用,在HBV感染的慢性本质中起主要作用。目前的目标治疗HBV感染的发展是一种临床结果称为功能性治愈,这标志着对感染的持续有效的免疫控制。虽然去除球形SVP(和他们携带的HBsAg)是实现功能治愈的重要里程碑,由于组装的不同机制,目前的疗法很少实现这一结果,分泌,和SVP的持久性,直接作用的抗病毒药物或免疫疗法的靶向性较差。在这篇评论中,目前对慢性HBV感染中球形SVP的产生和持续存在的不同机制及其免疫抑制活性的理解,以及目前正在开发的治疗方法,目标是清除球形SVP并实现功能性治愈.
    In diverse viral infections, the production of excess viral particles containing only viral glycoproteins (subviral particles or SVP) is commonly observed and is a commonly evolved mechanism for immune evasion. In hepatitis B virus (HBV) infection, spherical particles contain the hepatitis B surface antigen, outnumber infectious virus 10 000-100 000 to 1, and have diverse inhibitory effects on the innate and adaptive immune response, playing a major role in the chronic nature of HBV infection. The current goal of therapies in development for HBV infection is a clinical outcome called functional cure, which signals a persistent and effective immune control of the infection. Although removal of spherical SVP (and the HBsAg they carry) is an important milestone in achieving functional cure, this outcome is rarely achieved with current therapies due to distinct mechanisms for assembly, secretion, and persistence of SVP, which are poorly targeted by direct acting antivirals or immunotherapies. In this Review, the current understanding of the distinct mechanisms involved in the production and persistence of spherical SVP in chronic HBV infection and their immunoinhibitory activity will be reviewed as well as current therapies in development with the goal of clearing spherical SVP and achieving functional cure.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    乙型肝炎病毒(HBV)共价闭合环状DNA(cccDNA)的存在和HBVDNA永久整合到宿主基因组中赋予病毒再激活和肝细胞癌的风险。单独的核苷/核苷酸类似物很少或没有能力消除由cccDNA或整合的HBVDNA组成的复制型HBV模板。最近,CRISPR/Cas9技术作为一种很有前景的基因组编辑工具得到了广泛的应用,和HBV特异性CRISPR-Cas9系统显示有效介导HBVcccDNA破坏。然而,整合的HBVDNA片段被认为是重要的致癌特性,它是病毒在稳定的HBV细胞系中复制和表达的重要模板。在这项研究中,我们在稳定的HBV细胞系中完全切除了全长3,175bp的整合HBVDNA片段,并破坏了HBVcccDNA。在HBV切除的细胞系中,细胞内的HBVcccDNA,HBV上清液DNA,HBsAg,和HBeAg保持低于负临界值超过10个月。此外,通过全基因组测序,我们分析了脱靶效应并排除了细胞污染.这是首次在稳定的HBV细胞系中完全根除HBV感染。这些发现表明,CRISPR-Cas9系统是一种潜在的强大工具,能够促进彻底或“无菌”HBV治愈。
    The presence of hepatitis B virus (HBV) covalently closed circular DNA (cccDNA) and the permanent integration of HBV DNA into the host genome confers the risk of viral reactivation and hepatocellular carcinoma. Nucleoside/nucleotide analogs alone have little or no capacity to eliminate replicative HBV templates consisting of cccDNA or integrated HBV DNA. Recently, CRISPR/Cas9 technology has been widely applied as a promising genome-editing tool, and HBV-specific CRISPR-Cas9 systems were shown to effectively mediate HBV cccDNA disruption. However, the integrated HBV DNA fragments are considered as important pro-oncogenic properties and it serves as an important template for viral replication and expression in stable HBV cell line. In this study, we completely excised a full-length 3,175-bp integrated HBV DNA fragment and disrupted HBV cccDNA in a stable HBV cell line. In HBV-excised cell line, the HBV cccDNA inside cells, supernatant HBV DNA, HBsAg, and HBeAg remained below the negative critical values for more than 10 months. Besides, by whole genome sequencing, we analyzed off-target effects and excluded cell contamination. It is the first time that the HBV infection has been fully eradicated in a stable HBV cell line. These findings demonstrate that the CRISPR-Cas9 system is a potentially powerful tool capable of promoting a radical or \"sterile\" HBV cure.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号