Cell Transformation, Viral

  • 文章类型: Journal Article
    背景:默克尔细胞癌(MCC)是一种侵袭性皮肤癌,比黑色素瘤致命三倍。2008年,发现80%的MCC病例是由一种新型多瘤病毒的基因组整合引起的,默克尔细胞多瘤病毒(MCPyV),及其小的和截短的大的肿瘤抗原的表达(ST和LT-t,分别)。MCPyV属于人类多瘤病毒家族;然而,它是唯一与癌症有明确关联的。
    方法:为了研究各种多瘤病毒肿瘤抗原在细胞转化中的作用和机制,用pLENTIMCPyVLT-t转导大鼠2和293A细胞,MCPyVST,TSPyVST,HPyV7ST,或空pLENTI并通过多种转化测定进行评估,和亚细胞分裂。使用单因素方差分析检验来评估统计学显著性。
    结果:软琼脂,扩散,倍增时间,葡萄糖摄取,和血清依赖性测定证实ST是MCPyV的主要转化蛋白。此外,发现MCPyVST是独特的转化,与其他非癌性人多瘤病毒的ST抗原一样,在进行类似评估时,丝裂体发育不全的脊髓相关多瘤病毒(TSPyV)和人多瘤病毒7(HPyV7)没有转化.转化和非转化肿瘤抗原之间的结构差异的鉴定揭示MCPyVST的独特转化结构域可能位于MCPyVST独特区域的结构上不同的环内。在所有已知的MCPyVST细胞相互作用器中,62%是完全或暂时的核,表明尽管没有经典的核定位信号,但MCPyVST仍位于细胞核。的确,亚细胞分馏证实,MCPyVST可以通过目前未知的,调节机制独立于其小尺寸,因为HPyV7和TSPyVST蛋白不能核易位。尽管发现核定位对MCPyVST的几种转化特性很重要,细胞质隔离的MCPyVST也具有一些特性,提示MCPyVST可能在各个亚细胞区室中执行不同的转化功能。
    结论:一起,这些数据进一步阐明了MCPyVST和其他多瘤病毒ST蛋白之间的独特差异,这对于理解MCPyV是唯一已知的人类致癌多瘤病毒是必要的.
    BACKGROUND: Merkel Cell Carcinoma (MCC) is an aggressive skin cancer that is three times deadlier than melanoma. In 2008, it was found that 80% of MCC cases are caused by the genomic integration of a novel polyomavirus, Merkel Cell Polyomavirus (MCPyV), and the expression of its small and truncated large tumor antigens (ST and LT-t, respectively). MCPyV belongs to a family of human polyomaviruses; however, it is the only one with a clear association to cancer.
    METHODS: To investigate the role and mechanisms of various polyomavirus tumor antigens in cellular transformation, Rat-2 and 293A cells were transduced with pLENTI MCPyV LT-t, MCPyV ST, TSPyV ST, HPyV7 ST, or empty pLENTI and assessed through multiple transformation assays, and subcellular fractionations. One-way ANOVA tests were used to assess statistical significance.
    RESULTS: Soft agar, proliferation, doubling time, glucose uptake, and serum dependence assays confirmed ST to be the dominant transforming protein of MCPyV. Furthermore, it was found that MCPyV ST is uniquely transforming, as the ST antigens of other non-oncogenic human polyomaviruses such as Trichodysplasia Spinulosa-Associated Polyomavirus (TSPyV) and Human Polyomavirus 7 (HPyV7) were not transforming when similarly assessed. Identification of structural dissimilarities between transforming and non-transforming tumor antigens revealed that the uniquely transforming domain(s) of MCPyV ST are likely located within the structurally dissimilar loops of the MCPyV ST unique region. Of all known MCPyV ST cellular interactors, 62% are exclusively or transiently nuclear, suggesting that MCPyV ST localizes to the nucleus despite the absence of a canonical nuclear localization signal. Indeed, subcellular fractionations confirmed that MCPyV ST could achieve nuclear localization through a currently unknown, regulated mechanism independent of its small size, as HPyV7 and TSPyV ST proteins were incapable of nuclear translocation. Although nuclear localization was found to be important for several transforming properties of MCPyV ST, some properties were also performed by a cytoplasmic sequestered MCPyV ST, suggesting that MCPyV ST may perform different transforming functions in individual subcellular compartments.
    CONCLUSIONS: Together, these data further elucidate the unique differences between MCPyV ST and other polyomavirus ST proteins necessary to understand MCPyV as the only known human oncogenic polyomavirus.
    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
    B细胞感染后,EB病毒(EBV)参与介导细胞增殖和转化的宿主途径,有助于病毒驱动免疫失调和淋巴生成的倾向。我们发现EBV蛋白EBNA2通过驱动代谢酶IDO1在感染的B细胞中的表达来启动NAD从头生物合成。病毒强制NAD产生持续的线粒体复合物I活性,使ATP生产与增殖和转化的生物能量需求相匹配。在移植患者中,IDO1在EBV感染的B细胞中的表达,和IDO1活性增加的血清特征,早于淋巴瘤的发展。在感染EBV的人源化小鼠中,IDO1抑制降低病毒血症和淋巴生成。病毒精心策划的NAD生物合成是,因此,EBV驱动的B细胞转化的可药用代谢脆弱性打开EBV相关疾病的治疗可能性。
    After infection of B cells, Epstein-Barr virus (EBV) engages host pathways that mediate cell proliferation and transformation, contributing to the propensity of the virus to drive immune dysregulation and lymphomagenesis. We found that the EBV protein EBNA2 initiates nicotinamide adenine dinucleotide (NAD) de novo biosynthesis by driving expression of the metabolic enzyme indoleamine 2,3-dioxygenase 1 (IDO1) in infected B cells. Virus-enforced NAD production sustained mitochondrial complex I activity, to match adenosine triphosphate (ATP) production with bioenergetic requirements of proliferation and transformation. In transplant patients, IDO1 expression in EBV-infected B cells, and a serum signature of increased IDO1 activity, preceded development of lymphoma. In humanized mice infected with EBV, IDO1 inhibition reduced both viremia and lymphomagenesis. Virus-orchestrated NAD biosynthesis is therefore a druggable metabolic vulnerability of EBV-driven B cell transformation, opening therapeutic possibilities for EBV-related diseases.
    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
    EB病毒(EBV)潜伏膜蛋白1(LMP1)驱动病毒B细胞转化和肿瘤发生。LMP1的转化活性取决于其C端激活区2(CTAR2),通过参与TNF受体相关因子6(TRAF6)诱导NF-κB和JNK。TRAF6募集到LMP1的机制及其在LMP1信号传导中的作用仍然难以捉摸。在这里,我们证明了TRAF6与CTAR2内的病毒TRAF6结合基序直接相互作用。由分子建模支持的功能和NMR研究提供了对LMP1-TRAF6复合物结构的洞察,与CD40-TRAF6不同。TRAF6对LMP1的直接募集对于CTAR2激活NF-κB和LMP1驱动的淋巴瘤的存活至关重要。抑制肽对LMP1-TRAF6复合物的破坏干扰了EBV转化的B细胞的存活。在这项工作中,我们将LMP1-TRAF6确定为关键的病毒-宿主界面,并验证了这种相互作用作为EBV相关癌症的潜在治疗靶点.
    Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) drives viral B cell transformation and oncogenesis. LMP1\'s transforming activity depends on its C-terminal activation region 2 (CTAR2), which induces NF-κB and JNK by engaging TNF receptor-associated factor 6 (TRAF6). The mechanism of TRAF6 recruitment to LMP1 and its role in LMP1 signalling remains elusive. Here we demonstrate that TRAF6 interacts directly with a viral TRAF6 binding motif within CTAR2. Functional and NMR studies supported by molecular modeling provide insight into the architecture of the LMP1-TRAF6 complex, which differs from that of CD40-TRAF6. The direct recruitment of TRAF6 to LMP1 is essential for NF-κB activation by CTAR2 and the survival of LMP1-driven lymphoma. Disruption of the LMP1-TRAF6 complex by inhibitory peptides interferes with the survival of EBV-transformed B cells. In this work, we identify LMP1-TRAF6 as a critical virus-host interface and validate this interaction as a potential therapeutic target in EBV-associated cancer.
    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
    重要的是EB病毒(EBV)有助于伯基特淋巴瘤和移植后淋巴增生性疾病(PTLD)。EBV转化程序激活脂质代谢,将B细胞转化为永生化的淋巴母细胞系(LCL),PTLD模型。我们发现EBV转化的阶段会产生不同程度的脂质活性氧(ROS)副产物,B细胞生长的Burkitt样阶段需要谷胱甘肽过氧化物酶4及其辅因子谷胱甘肽进行脂质ROS解毒。在转化的早期阶段或伯基特细胞中,这种氧化还原防御的扰动引发了铁凋亡,程序性细胞死亡途径。LCL不太依赖这种防御,与EBV延迟程序有关的区别。这突出了铁凋亡诱导作为预防或治疗某些EBV+淋巴瘤的潜在治疗方法。
    SignificanceEpstein-Barr virus (EBV) contributes to Burkitt lymphoma and post-transplant lymphoproliferative disease (PTLD). EBV-transforming programs activate lipid metabolism to convert B cells into immortalized lymphoblastoid cell lines (LCL), a PTLD model. We found that stages of EBV transformation generate lipid reactive oxygen species (ROS) byproducts to varying degrees, and that a Burkitt-like phase of B cell outgrowth requires lipid ROS detoxification by glutathione peroxidase 4 and its cofactor glutathione. Perturbation of this redox defense in early stages of transformation or in Burkitt cells triggered ferroptosis, a programmed cell death pathway. LCLs were less dependent on this defense, a distinction tied to EBV latency programs. This highlights ferroptosis induction as a potential therapeutic approach for prevention or treatment of certain EBV+ lymphomas.
    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
    人乳头瘤病毒(HPV)是全球最常见的性传播因子。早期预防HPV疫苗接种是预防这种疾病的安全有效方法。HPV疫苗提供了针对几种致癌HPV毒株的更多保护。三种预防性HPV疫苗已被批准针对高危HPV类型并预防HPV相关疾病。这些现有的疫苗基于重组DNA技术和被组装以形成HPV空壳的纯化的L1蛋白。预防性疫苗是高度免疫原性的并且可以诱导特异性中和抗体的产生。然而,治疗性疫苗不同于这些预防性疫苗。它们诱导了细胞介导的针对转化细胞的免疫,而不是中和抗体.第二代预防性HPV疫苗,使用具有成本效益的生产策略由替代病毒成分制成,正在接受临床评估。本综述的目的是为读者提供对HPV疫苗类型以及每种疫苗的效率的完整和最新的综述。
    Human papillomavirus (HPV) is the most common sexually transmitted agent worldwide. Early prevention with HPV vaccination is a safe and effective method against this disease. HPV vaccines provided more protection against several oncogenic HPV strains. Three prophylactic HPV vaccines have been approved to target high-risk HPV types and protect against HPV-related disorders. These existing vaccines are based on the recombinant DNA technology and purified L1 protein that is assembled to form HPV empty shells. The prophylactic vaccines are highly immunogenic and can induce production of specific neutralizing antibodies. However, therapeutic vaccines are different from these prophylactic vaccines. They induced cell-mediated immunity against transformed cells, instead of neutralizing antibodies. The second generation of prophylactic HPV vaccines, made from alternative viral components using cost-effective production strategies, is undergoing clinical evaluation. The purpose of this review is to provide a complete and up-to-date review of the types of HPV vaccines and the efficiency of each of them for readers.
    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
    人类T细胞白血病病毒1型(HTLV-1)是一种致癌逆转录病毒,是成人T细胞白血病/淋巴瘤(ATL)的致病传染因子,侵袭性和致命的CD4+T细胞恶性肿瘤,和HTLV-1相关的脊髓病/热带痉挛性轻瘫(HAM/TSP),慢性神经系统疾病.感染个体中的疾病进展是HTLV-1驱动的CD4+T细胞的克隆扩增的结果,并且通常与病毒癌蛋白Tax和Hbz的活性相关。密切相关的病毒,HTLV-2表现出相似的基因组特征和转化T细胞的能力,但是非致病性的。体外,HTLV-1主要使CD4+T细胞永生化或转化,而HTLV-2对CD8+T细胞表现出转化嗜性。这种独特的向性在感染者中得到了概括。通过比较研究,HTLV-1/2的这种不同向性的遗传决定子已被定位到病毒包膜(Env)。在这次审查中,我们探讨了Env在初始病毒进入后的新作用,并研究了其对HTLV-1介导的疾病发展的贡献的当前观点.
    Human T-cell leukemia virus type 1 (HTLV-1) is an oncogenic retrovirus that is the causative infectious agent of adult T-cell leukemia/lymphoma (ATL), an aggressive and fatal CD4+ T-cell malignancy, and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP), a chronic neurological disease. Disease progression in infected individuals is the result of HTLV-1-driven clonal expansion of CD4+ T-cells and is generally associated with the activities of the viral oncoproteins Tax and Hbz. A closely related virus, HTLV-2, exhibits similar genomic features and the capacity to transform T-cells, but is non-pathogenic. In vitro, HTLV-1 primarily immortalizes or transforms CD4+ T-cells, while HTLV-2 displays a transformation tropism for CD8+ T-cells. This distinct tropism is recapitulated in infected people. Through comparative studies, the genetic determinant for this divergent tropism of HTLV-1/2 has been mapped to the viral envelope (Env). In this review, we explore the emerging roles for Env beyond initial viral entry and examine current perspectives on its contributions to HTLV-1-mediated disease development.
    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
    多功能腺病毒E1B-55K磷蛋白是病毒复制的主要调节因子,在病毒介导的细胞转化中起着关键作用。虽然人们对其在致癌细胞转化中的功能了解很多,E1B-55K参与病毒基因表达调控的基本特征和确切机制尚不清楚.因此,这项工作旨在揭示使用野生型人腺病毒C5(HAdV-C5)E1B-55K调节病毒mRNA生物发生的基本核内原理,E1B-55K表达被取消的病毒突变体,和表达磷模拟物E1B-55K的突变体。通过亚核分馏,mRNADNA,和蛋白质分析以及荧光素酶报告基因测定,我们显示(i)E1B-55K促进病毒晚期mRNA从其在病毒复制区室(RCs)的合成位点有效释放到周围的核质,(ii)E1B-55K调节RCs中病毒基因转录和剪接的速率,(iii)E1B-55K参与病毒基因表达的时间调节,(iv)E1B-55K可以增强或抑制病毒早期和晚期启动子的表达,和(v)E1B-55K的磷酸化调节蛋白质对这些活性中的每一种的时间效应。一起,这些数据表明E1B-55K是HAdV-C5感染期间病毒基因的磷酸化依赖性转录和转录后调节因子.重要性人腺病毒是研究基因表达和剪接的基本方面的有用模型。此外,它们是临床上最常用的病毒载体之一。然而,通常在腺病毒载体中修饰的必需病毒蛋白活性的关键方面尚未得到充分描述。一个突出的例子是多功能腺病毒癌蛋白E1B-55K,其已知促进有效的病毒基因组复制和表达,同时抑制宿主基因表达和抗病毒宿主应答。我们的研究结合了不同的定量方法来研究E1B-55K如何促进病毒mRNA的生物发生。这里提供的数据提出了E1B-55K作为病毒基因的磷酸化依赖性转录和转录后调节因子的新作用。
    The multifunctional adenoviral E1B-55K phosphoprotein is a major regulator of viral replication and plays key roles in virus-mediated cell transformation. While much is known about its function in oncogenic cell transformation, the underlying features and exact mechanisms that implicate E1B-55K in the regulation of viral gene expression are less well understood. Therefore, this work aimed to unravel basic intranuclear principles of E1B-55K-regulated viral mRNA biogenesis using wild-type human adenovirus C5 (HAdV-C5) E1B-55K, a virus mutant with abrogated E1B-55K expression, and a mutant that expresses a phosphomimetic E1B-55K. By subnuclear fractionation, mRNA, DNA, and protein analyses as well as luciferase reporter assays, we show that (i) E1B-55K promotes the efficient release of viral late mRNAs from their site of synthesis in viral replication compartments (RCs) to the surrounding nucleoplasm, (ii) E1B-55K modulates the rate of viral gene transcription and splicing in RCs, (iii) E1B-55K participates in the temporal regulation of viral gene expression, (iv) E1B-55K can enhance or repress the expression of viral early and late promoters, and (v) the phosphorylation of E1B-55K regulates the temporal effect of the protein on each of these activities. Together, these data demonstrate that E1B-55K is a phosphorylation-dependent transcriptional and posttranscriptional regulator of viral genes during HAdV-C5 infection. IMPORTANCE Human adenoviruses are useful models to study basic aspects of gene expression and splicing. Moreover, they are one of the most commonly used viral vectors for clinical applications. However, key aspects of the activities of essential viral proteins that are commonly modified in adenoviral vectors have not been fully described. A prominent example is the multifunctional adenoviral oncoprotein E1B-55K that is known to promote efficient viral genome replication and expression while simultaneously repressing host gene expression and antiviral host responses. Our study combined different quantitative methods to study how E1B-55K promotes viral mRNA biogenesis. The data presented here propose a novel role for E1B-55K as a phosphorylation-dependent transcriptional and posttranscriptional regulator of viral genes.
    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
    卡波西肉瘤相关疱疹病毒(KSHV)感染与多种癌症有因果关系。KSHV诱导肿瘤发生的机制尚不清楚。通过在KSHV诱导的原代细胞细胞转化模型中进行CRISPR-Cas9筛选,我们确定了KSHV诱导的细胞转化所必需的表观遗传调节因子.检查前9个基因的TCGA数据集,包括含1的富含谷氨酸的WD重复序列(GRWD1),由KSHV上调的WD40家族蛋白,对KSHV转化细胞(KMM)的细胞增殖和存活有积极影响,但对匹配的原代细胞(MM)没有影响,揭示了它们的表达对许多类型癌症患者生存的预测价值。我们揭示了与MM细胞相比,KMM细胞中包括H3K4me3表观遗传活性标记的全球表观遗传重塑。敲除GRWD1抑制细胞增殖,细胞转化,和肿瘤形成并引起KMM细胞中整体H3K4me3标记的下调。GRWD1与WD重复结构域5(WDR5)相互作用,H3K4甲基转移酶复合物的核心蛋白,和几种H3K4me3甲基转移酶,包括髓系白血病2(MLL2)。WDR5和MLL2表型敲除GRWD1敲除,导致全球H3K4me3标记减少,并改变了相似基因的表达。转录组测序(RNA-seq)和染色质免疫沉淀测序(ChIP-seq)分析进一步鉴定了由GRWD1,WDR5和MLL2调节的常见和不同的细胞基因和途径。这些结果表明,KSHV劫持GRWD1-WDR5-MLL2表观遗传复合物来调节特定基因的H3K4me3甲基化,这对于KSHV诱导的细胞转化至关重要。我们的工作已经确定了表观遗传复合物作为KSHV诱导的癌症的新治疗靶标。重要性通过进行全基因组CRISPR-Cas9筛选,我们已经确定了KSHV诱导的细胞转化所必需的细胞表观遗传调节因子.其中,GRWD1通过与WDR5和MLL2相互作用并将它们招募到KSHV转化细胞中特定基因的染色质基因座来调节表观遗传活性标记H3K4me3。因此,KSHV劫持GRWD1-WDR5-MLL2复合物以重塑细胞表观基因组并诱导细胞转化。由于GRWD1的失调与几种癌症的不良预后有关,GRWD1也可能是其他病毒性或非病毒性癌症的关键驱动因素。
    Infection by Kaposi\'s sarcoma-associated herpesvirus (KSHV) is causally associated with numerous cancers. The mechanism of KSHV-induced oncogenesis remains unclear. By performing a CRISPR-Cas9 screening in a model of KSHV-induced cellular transformation of primary cells, we identified epigenetic regulators that were essential for KSHV-induced cellular transformation. Examination of TCGA data sets of the top 9 genes, including glutamate-rich WD repeat containing 1 (GRWD1), a WD40 family protein upregulated by KSHV, that had positive effects on cell proliferation and survival of KSHV-transformed cells (KMM) but not the matched primary cells (MM), uncovered the predictive values of their expressions for patient survival in numerous types of cancer. We revealed global epigenetic remodeling including H3K4me3 epigenetic active mark in KMM cells compared to MM cells. Knockdown of GRWD1 inhibited cell proliferation, cellular transformation, and tumor formation and caused downregulation of global H3K4me3 mark in KMM cells. GRWD1 interacted with WD repeat domain 5 (WDR5), the core protein of H3K4 methyltransferase complex, and several H3K4me3 methyltransferases, including myeloid leukemia 2 (MLL2). Knockdown of WDR5 and MLL2 phenocopied GRWD1 knockdown, caused global reduction of H3K4me3 mark, and altered the expression of similar sets of genes. Transcriptome sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) analyses further identified common and distinct cellular genes and pathways that were regulated by GRWD1, WDR5, and MLL2. These results indicate that KSHV hijacks the GRWD1-WDR5-MLL2 epigenetic complex to regulate H3K4me3 methylation of specific genes, which is essential for KSHV-induced cellular transformation. Our work has identified an epigenetic complex as a novel therapeutic target for KSHV-induced cancers. IMPORTANCE By performing a genome-wide CRISPR-Cas9 screening, we have identified cellular epigenetic regulators that are essential for KSHV-induced cellular transformation. Among them, GRWD1 regulates epigenetic active mark H3K4me3 by interacting with WDR5 and MLL2 and recruiting them to chromatin loci of specific genes in KSHV-transformed cells. Hence, KSHV hijacks the GRWD1-WDR5-MLL2 complex to remodel cellular epigenome and induce cellular transformation. Since the dysregulation of GRWD1 is associated with poor prognosis in several types of cancer, GRWD1 might also be a critical driver in other viral or nonviral cancers.
    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
    Human T cell leukemia virus type 1 (HTLV-1) mainly infects CD4+ T cells and induces chronic, persistent infection in infected individuals, with some developing adult T cell leukemia/lymphoma (ATL). HTLV-1 alters cellular differentiation, activation, and survival; however, it is unknown whether and how these changes contribute to the malignant transformation of infected cells. In this study, we used single-cell RNA-sequencing and T cell receptor-sequencing to investigate the differentiation and HTLV-1-mediated transformation of T cells. We analyzed 87,742 PBMCs from 12 infected and 3 uninfected individuals. Using multiple independent bioinformatics methods, we demonstrated the seamless transition of naive T cells into activated T cells, whereby HTLV-1-infected cells in an activated state further transformed into ATL cells, which are characterized as clonally expanded, highly activated T cells. Notably, the greater the activation state of ATL cells, the more they acquire Treg signatures. Intriguingly, the expression of HLA class II genes in HTLV-1-infected cells was uniquely induced by the viral protein Tax and further upregulated in ATL cells. Functional assays revealed that HTLV-1-infected cells upregulated HLA class II molecules and acted as tolerogenic antigen-presenting cells to induce anergy of antigen-specific T cells. In conclusion, our study revealed the in vivo mechanisms of HTLV-1-mediated transformation and immune escape at the single-cell level.
    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
    Papillomavirus L1 and L2, the major and minor capsid proteins, play significant roles in viral assembly, entry, and propagation. In the current study, we investigate the impact of L1 and L2 on viral life cycle and tumor growth with a newly established mouse papillomavirus (MmuPV1) infection model. MmuPV1 L1 knockout, L2 knockout, and L1 plus L2 knockout mutant genomes (designated as L1ATGko-4m, L2ATGko, and L1-L2ATGko respectively) were generated. The mutants were examined for their ability to generate lesions in athymic nude mice. Viral activities were examined by qPCR, immunohistochemistry (IHC), in situ hybridization (ISH), and transmission electron microscopy (TEM) analyses. We demonstrated that viral DNA replication and tumor growth occurred at both cutaneous and mucosal sites infected with each of the mutants. Infections involving L1ATGko-4m, L2ATGko, and L1-L2ATGko mutant genomes generally resulted in smaller tumor sizes compared to infection with the wild type. The L1 protein was absent in L1ATGko-4m and L1-L2ATGko mutant-treated tissues, even though viral transcripts and E4 protein expression were robust. Therefore, L1 is not essential for MmuPV1-induced tumor growth, and this finding parallels our previous observations in the rabbit papillomavirus model. Very few viral particles were detected in L2ATGko mutant-infected tissues. Interestingly, the localization of L1 in lesions induced by L2ATGko was primarily cytoplasmic rather than nuclear. The findings support the hypothesis that the L2 gene influences the expression, location, transport, and assembly of the L1 protein in vivo.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号