Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.461
Filter
1.
Virol J ; 21(1): 125, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38831469

ABSTRACT

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.


Subject(s)
Antigens, Viral, Tumor , Cell Nucleus , Merkel cell polyomavirus , Merkel cell polyomavirus/genetics , Merkel cell polyomavirus/physiology , Humans , Antigens, Viral, Tumor/genetics , Antigens, Viral, Tumor/metabolism , Cell Nucleus/virology , Cell Nucleus/metabolism , Animals , Rats , Nuclear Localization Signals , Carcinoma, Merkel Cell/virology , Cell Line , Skin Neoplasms/virology , Skin Neoplasms/pathology , Cell Transformation, Viral , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Polyomavirus Infections/virology
2.
Nat Commun ; 15(1): 414, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38195569

ABSTRACT

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.


Subject(s)
Epstein-Barr Virus Infections , Lymphoma, B-Cell , Humans , Herpesvirus 4, Human , TNF Receptor-Associated Factor 6 , Epstein-Barr Virus Infections/complications , NF-kappa B , Cell Transformation, Neoplastic , Cell Transformation, Viral
3.
Proc Natl Acad Sci U S A ; 119(11): e2118300119, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35275790

ABSTRACT

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.


Subject(s)
B-Lymphocytes , Burkitt Lymphoma , Cell Transformation, Viral , Ferroptosis , Herpesvirus 4, Human , Virus Latency , B-Lymphocytes/immunology , B-Lymphocytes/virology , Burkitt Lymphoma/virology , Ferroptosis/immunology , Herpesvirus 4, Human/physiology , Humans , Lipid Metabolism , Lymphocyte Activation , Reactive Oxygen Species/metabolism
4.
Viruses ; 14(1)2022 01 13.
Article in English | MEDLINE | ID: mdl-35062342

ABSTRACT

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.


Subject(s)
Gene Products, env/genetics , Gene Products, env/metabolism , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/metabolism , Leukemia-Lymphoma, Adult T-Cell/virology , Virus Internalization , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Cell Transformation, Viral/genetics , Gene Products, tax/genetics , Human T-lymphotropic virus 1/pathogenicity , Human T-lymphotropic virus 2/genetics , Humans , Oncogenes
5.
J Virol ; 96(5): e0206221, 2022 03 09.
Article in English | MEDLINE | ID: mdl-35019711

ABSTRACT

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.


Subject(s)
Adenovirus Infections, Human , Adenoviruses, Human , Cell Transformation, Viral , Gene Expression Regulation, Viral , Viral Proteins , Adenovirus Infections, Human/physiopathology , Adenovirus Infections, Human/virology , Adenoviruses, Human/genetics , Adenoviruses, Human/metabolism , Cell Transformation, Viral/genetics , Humans , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Viral Proteins/metabolism
6.
Br J Haematol ; 196(3): 681-689, 2022 02.
Article in English | MEDLINE | ID: mdl-34617271

ABSTRACT

Sporadic Burkitt lymphoma (BL) is the most frequent tumour of children and adolescents but a rare subtype of lymphomas in adults. To date most molecular data have been obtained from lymphomas arising in the young. Recently, Epstein-Barr virus (EBV) positive and negative BL in young patients was shown to differ in molecular features. In the present study, we present a large age-overarching cohort of sporadic BL (n = 162) analysed by immunohistochemistry, translocations of MYC proto-oncogene, basic helix-loop-helix transcription factor (MYC), B-cell leukaemia/lymphoma 2 (BCL2) and B-cell leukaemia/lymphoma 6 (BCL6) and by targeted sequencing. We illustrate an age-associated inter-tumoral molecular heterogeneity in this disease. Mutations affecting inhibitor of DNA binding 3, HLH protein (ID3), transcription factor 3 (TCF3) and cyclin D3 (CCND3), which are highly recurrent in paediatric BL, and expression of sex determining region Y-box transcription factor 11 (SOX11) declined with patient age at diagnosis (P = 0·0204 and P = 0·0197 respectively). In contrast, EBV was more frequently detected in adult patients (P = 0·0262). Irrespective of age, EBV-positive sporadic BL showed significantly less frequent mutations in ID3/TCF3/CCND3 (P = 0·0088) but more often mutations of G protein subunit alpha 13 (GNA13; P = 0·0368) and forkhead box O1 (FOXO1; P = 0·0044) compared to EBV-negative tumours. Our findings suggest that among sporadic BL an EBV-positive subgroup of lymphomas increases with patient age that shows distinct pathogenic features reminiscent of EBV-positive endemic BL.


Subject(s)
Burkitt Lymphoma/epidemiology , Burkitt Lymphoma/etiology , Disease Susceptibility , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/physiology , Mutation , Adolescent , Adult , Age Factors , Age of Onset , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Burkitt Lymphoma/diagnosis , Cell Transformation, Viral , Child , Child, Preschool , DNA Mutational Analysis , Epstein-Barr Virus Infections/virology , Female , Gene Expression , Gene Expression Profiling , Genetic Predisposition to Disease , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Infant , Infant, Newborn , Male , Middle Aged , Young Adult
7.
J Cell Biochem ; 123(2): 161-182, 2022 02.
Article in English | MEDLINE | ID: mdl-34520596

ABSTRACT

Viruses are known to cause various diseases in human and also infect other species such as animal plants, fungi, and bacteria. Replication of viruses depends upon their interaction with hosts. Human cells are prone to such unwanted viral infections. Disintegration and reconstitution require host machinery and various macromolecules like DNA, RNA, and proteins are invaded by viral particles. E3 ubiquitin ligases are known for their specific function, that is, recognition of their respective substrates for intracellular degradation. Still, we do not understand how ubiquitin proteasome system-based enzymes E3 ubiquitin ligases do their functional interaction with different viruses. Whether E3 ubiquitin ligases help in the elimination of viral components or viruses utilize their molecular capabilities in their intracellular propagation is not clear. The first time our current article comprehends fundamental concepts and new insights on the different viruses and their interaction with various E3 Ubiquitin Ligases. In this review, we highlight the molecular pathomechanism of viruses linked with E3 Ubiquitin Ligases dependent mechanisms. An enhanced understanding of E3 Ubiquitin Ligase-mediated removal of viral proteins may open new therapeutic strategies against viral infections.


Subject(s)
Ubiquitin-Protein Ligases/physiology , Viral Proteins/physiology , Virus Diseases/enzymology , Virus Replication/physiology , Cell Transformation, Viral/physiology , Cullin Proteins/physiology , Endosomes/virology , Host-Pathogen Interactions , Humans , Immunity, Innate , Inflammation/enzymology , Inflammation/virology , Neoplasms/enzymology , Neoplasms/virology , Oncogenic Viruses/physiology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Tripartite Motif Proteins/physiology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Virus Diseases/immunology , Virus Diseases/virology , Virus Replication/drug effects , COVID-19 Drug Treatment
8.
Oral Dis ; 28(8): 2230-2238, 2022 Nov.
Article in English | MEDLINE | ID: mdl-34060687

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate and compare alterations in gene expression using two distinct immortalization methods (hTERT and HPV16-E6/E7) in ameloblastoma cell lines. MATERIALS AND METHODS: A primary cell culture derived from human ameloblastoma (AME-1) was established and immortalized by two different methods using a transfection processes to hTERT and HPV-E6/E7. The RNA-seq was used to verify which immortalization method had less influence on gene expression. It was performed in four steps: extraction and collection of mRNA, PCR amplification, comparison with the human reference genome, and analysis of differential expression. The genes with differentiated expression were identified and mapped. RESULTS: RNA-seq revealed genetic alterations in ameloblastoma cell lines after the immortalization process, including increased expression of tumor genes like MYC, E2F1, BRAF, HRAS, and HTERT, and a decrease in tumor suppressor genes like P53, P21, and Rb. CONCLUSIONS: It is possible to affirm that cell immortalization is not an inert method regarding gene regulation mechanisms and the hTERT method (AME-TERT) presented fewer changes in gene expression levels.


Subject(s)
Ameloblastoma , Oncogene Proteins, Viral , Humans , Ameloblastoma/genetics , Cell Line , Cell Transformation, Viral/genetics , Gene Expression , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomaviridae/genetics , Papillomavirus E7 Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , RNA, Messenger , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
J Clin Invest ; 131(24)2021 12 15.
Article in English | MEDLINE | ID: mdl-34907908

ABSTRACT

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.


Subject(s)
Cell Transformation, Viral/immunology , Human T-lymphotropic virus 1/immunology , Leukemia-Lymphoma, Adult T-Cell/immunology , Lymphocyte Activation , T-Lymphocytes/immunology , Female , Gene Products, tax/immunology , HLA Antigens/immunology , Humans , Leukemia-Lymphoma, Adult T-Cell/virology , Male
10.
mBio ; 12(6): e0343121, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34933446

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Cell Transformation, Viral , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Herpesvirus 8, Human/physiology , Histones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Sarcoma, Kaposi/metabolism , Animals , Carrier Proteins/genetics , DNA-Binding Proteins/genetics , Herpesvirus 8, Human/genetics , Histones/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Nude , Neoplasm Proteins/genetics , Protein Binding , Sarcoma, Kaposi/enzymology , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/virology
11.
Cancer Sci ; 112(12): 5088-5099, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34609775

ABSTRACT

Epstein-Barr virus-associated lymphoproliferative disease (EBV-LPD) is frequently fatal. Innate immunity plays a key role in protecting against pathogens and cancers. The stimulator of interferon genes (STING) is regarded as a key adaptor protein allowing DNA sensors recognizing exogenous cytosolic DNA to activate the type I interferon signaling cascade. In terms of EBV tumorigenicity, the role of STING remains elusive. Here we showed that treatment with the STING inhibitor, C-176, suppressed EBV-induced transformation in peripheral blood mononuclear cells. In an EBV-LPD mouse model, C-176 treatment also inhibited tumor formation and prolonged survival. Treatment with B cells alone did not affect EBV transformation, but suppression of EBV-induced transformation was observed in the presence of T cells. Even without direct B cell-T cell contact in a transwell system, the inhibitor reduced the transformation activity, indicating that intercellular communication by humoral factors was critical to prevent EBV-induced transformation. These findings suggest that inhibition of STING signaling pathway with C-176 could be a new therapeutic target of EBV-LPD.


Subject(s)
Antineoplastic Agents/administration & dosage , Cell Transformation, Viral/drug effects , Epstein-Barr Virus Infections/drug therapy , Lymphoma, B-Cell/prevention & control , Membrane Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Epstein-Barr Virus Infections/immunology , HEK293 Cells , Herpesvirus 4, Human , Humans , Jurkat Cells , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/virology , Mice , Survival Analysis , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
12.
PLoS Pathog ; 17(9): e1009847, 2021 09.
Article in English | MEDLINE | ID: mdl-34492084

ABSTRACT

Increasing evidence suggests that Kaposi's sarcoma (KS) arises from Kaposi's sarcoma-associated herpesvirus (KSHV)-infected mesenchymal stem cells (MSCs) through mesenchymal-to-endothelial transition (MEndT). KSHV infection promotes MSC differentiation of endothelial lineage and acquisition of tumorigeneic phenotypes. To understand how KSHV induces MEndT and transforms MSCs to KS cells, we investigated the mechanism underlying KSHV-mediated MSC endothelial lineage differentiation. Like embryonic stem cells, MSC differentiation and fate determination are under epigenetic control. Prospero homeobox 1 (PROX1) is a master regulator that controls lymphatic vessel development and endothelial differentiation. We found that the PROX1 gene in MSCs harbors a distinctive bivalent epigenetic signature consisting of both active marker H3K4me3 and repressive marker H3K27me3, which poises expression of the genes, allowing timely activation upon differentiation signals or environmental stimuli. KSHV infection effectively resolves the bivalent chromatin by decreasing H3K27me3 and increasing H3K4me3 to activate the PROX1 gene. vIL-6 signaling leads to the recruitment of MLL2 and SET1 complexes to the PROX1 promoter to increase H3K4me3, and the vGPCR-VEGF-A axis is responsible for removing PRC2 from the promoter to reduce H3K27me3. Therefore, through a dual signaling process, KSHV activates PROX1 gene expression and initiates MEndT, which renders MSC tumorigenic features including angiogenesis, invasion and migration.


Subject(s)
Cell Differentiation/physiology , Cell Transformation, Viral/physiology , Homeodomain Proteins/metabolism , Mesenchymal Stem Cells/virology , Sarcoma, Kaposi/virology , Tumor Suppressor Proteins/metabolism , Gene Expression Regulation , Herpesvirus 8, Human , Humans
13.
Viruses ; 13(9)2021 09 14.
Article in English | MEDLINE | ID: mdl-34578405

ABSTRACT

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.


Subject(s)
Capsid Proteins/physiology , Mucous Membrane/virology , Oncogene Proteins, Viral/physiology , Papillomaviridae/physiology , Skin/virology , Animals , Capsid Proteins/genetics , Cell Transformation, Viral , DNA, Viral/biosynthesis , Female , Genome, Viral , Mice , Mice, Nude , Mutation , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Papillomaviridae/pathogenicity , Virus Replication
14.
mBio ; 12(5): e0109721, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34488443

ABSTRACT

The Epstein-Barr virus (EBV) protein LMP1 serves as a paradigm that engages complicated ubiquitination-mediated mechanisms to activate multiple transcription factors. p62 is a ubiquitin sensor and a signal-transducing adaptor that has multiple functions in diverse contexts. However, the interaction between p62 and oncogenic viruses is poorly understood. We recently reported a crucial role for p62 in oncovirus-mediated oxidative stress by acting as a selective autophagy receptor. In this following pursuit, we further discovered that p62 is upregulated in EBV type 3 compared to type 1 latency, with a significant contribution from NF-κB and AP1 activities downstream of LMP1 signaling. In turn, p62 participates in LMP1 signal transduction through its interaction with TRAF6, promoting TRAF6 ubiquitination and activation. As expected, short hairpin RNA (shRNA)-mediated knockdown (KD) of p62 transcripts reduces LMP1-TRAF6 interaction and TRAF6 ubiquitination, as well as p65 nuclear translocation, which was assessed by Amnis imaging flow cytometry. Strikingly, LMP1-stimulated NF-κB, AP1, and Akt activities are all markedly reduced in p62-/- mouse embryo fibroblasts (MEFs) and in EBV-negative Burkitt's lymphoma (BL) cell lines with CRISPR-mediated knockout (KO) of the p62-encoding gene. However, EBV-positive BL cell lines (type 3 latency) with CRISPR-mediated KO of the p62-encoding gene failed to survive. In consequence, shRNA-mediated p62 KD impairs the ability of LMP1 to regulate its target gene expression, promotes etoposide-induced apoptosis, and reduces the proliferation of lymphoblastic cell lines (LCLs). These important findings have revealed a previously unrecognized novel role for p62 in EBV latency and oncogenesis, which advances our understanding of the mechanism underlying virus-mediated oncogenesis. IMPORTANCE As a ubiquitin sensor and a signal-transducing adaptor, p62 is crucial for NF-κB activation, which involves the ubiquitin machinery, in diverse contexts. However, whether p62 is required for EBV LMP1 activation of NF-κB is an open question. In this study, we provide evidence that p62 is upregulated in EBV type 3 latency and, in turn, p62 mediates LMP1 signal transduction to NF-κB, AP1, and Akt by promoting TRAF6 ubiquitination and activation. In consequence, p62 deficiency negatively regulates LMP1-mediated gene expression, promotes etoposide-induced apoptosis, and reduces the proliferation of LCLs. These important findings identified p62 as a novel signaling component of the key viral oncogenic signaling pathway.


Subject(s)
Gene Expression Regulation , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , NF-kappa B/metabolism , Sequestosome-1 Protein/metabolism , Viral Matrix Proteins/genetics , Apoptosis , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Viral/genetics , Humans , Sequestosome-1 Protein/genetics , Signal Transduction , Viral Matrix Proteins/metabolism , Virus Latency
15.
Viruses ; 13(8)2021 07 28.
Article in English | MEDLINE | ID: mdl-34452344

ABSTRACT

Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV) independently cause human cancers, and both are maintained as plasmids in tumor cells. They differ, however, in their mechanisms of segregation; EBV partitions its genomes quasi-faithfully, while KSHV often clusters its genomes and partitions them randomly. Both viruses can infect the same B-cell to transform it in vitro and to cause primary effusion lymphomas (PELs) in vivo. We have developed simulations based on our measurements of these replicons in B-cells transformed in vitro to elucidate the synthesis and partitioning of these two viral genomes when in the same cell. These simulations successfully capture the biology of EBV and KSHV in PELs. They have revealed that EBV and KSHV replicate and partition independently, that they both contribute selective advantages to their host cell, and that KSHV pays a penalty to cluster its genomes.


Subject(s)
B-Lymphocytes/virology , Cell Transformation, Viral , Coinfection/virology , Epstein-Barr Virus Infections/virology , Herpesviridae Infections/virology , Herpesvirus 4, Human/physiology , Herpesvirus 8, Human/physiology , Herpesvirus 4, Human/genetics , Herpesvirus 8, Human/genetics , Humans , Lymphoma, Primary Effusion/virology , Virus Replication
16.
Int J Mol Sci ; 22(12)2021 Jun 18.
Article in English | MEDLINE | ID: mdl-34207106

ABSTRACT

Infection with human papillomavirus type 16 (HPV16) is one of the major risk factors for the development of cervical cancer. Our previous studies have demonstrated the involvement of the early oncoprotein E5 of HPV16 (16E5) in the altered isoform switch of fibroblast growth factor receptor 2 (FGFR2) and the consequent expression in human keratinocytes of the mesenchymal FGFR2c isoform, whose aberrant signaling leads to EMT, invasiveness, and dysregulated differentiation. Here, we aimed to establish the possible direct link between these pathological features or the appearance of FGFR2c and the expression of 16E5 in low-grade squamous intraepithelial lesions (LSILs). Molecular analysis showed that the FGFR2c expression displayed a statistically significant positive correlation with that of the viral oncoprotein, whereas the expression values of the epithelial FGR2b variant, as well as those of the differentiation markers keratin 10 (K10), loricrin (LOR) and involucrin (INV), were inversely linked to the 16E5 expression. In contrast, the expression of EMT-related transcription factors Snail1 and ZEB1 overlapped with that of 16E5, becoming a statistically significant positive correlation in the case of Snail2. Parallel analysis performed in human cervical LSIL-derived W12 cells, containing episomal HPV16, revealed that the depletion of 16E5 by siRNA was able to counteract these molecular events, proving to represent an effective strategy to identify the specific role of this viral oncoprotein in determining LSIL oncogenic and more aggressive profiles. Overall, coupling in vitro approaches to the molecular transcript analysis in ectocervical early lesions could significantly contribute to the characterization of specific gene expression profiles prognostic for those LSILs with a greater probability of direct neoplastic progression.


Subject(s)
Epithelial-Mesenchymal Transition , Gene Expression , Human papillomavirus 16/genetics , Oncogene Proteins, Viral/genetics , Papillomavirus Infections/complications , Uterine Cervical Neoplasms/etiology , Uterine Cervical Neoplasms/pathology , Cell Line , Cell Transformation, Viral , Cervix Uteri/pathology , Cervix Uteri/virology , Disease Susceptibility , Epithelial Cells/metabolism , Female , Humans , Keratinocytes/metabolism , Neoplasm Grading , Papillomavirus Infections/virology , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Uterine Cervical Neoplasms/metabolism
17.
Viruses ; 13(6)2021 06 11.
Article in English | MEDLINE | ID: mdl-34208172

ABSTRACT

The envelope of hepatitis B virus (HBV), which is required for the entry to hepatocytes, consists of a lipid bilayer derived from hepatocyte and HBV envelope proteins, large/middle/small hepatitis B surface antigen (L/M/SHBs). The mechanisms and host factors for the envelope formation in the hepatocytes are being revealed. HBV-infected hepatocytes release a large amount of subviral particles (SVPs) containing L/M/SHBs that facilitate escape from the immune system. Recently, novel drugs inhibiting the functions of the viral envelope and those inhibiting the release of SVPs have been reported. LHBs that accumulate in ER is considered to promote carcinogenesis and, especially, deletion mutants in the preS1/S2 domain have been reported to be associated with the development of hepatocellular carcinoma (HCC). In this review, we summarize recent reports on the findings regarding the biological characteristics of HBV envelope proteins, their involvement in HCC development and new agents targeting the envelope.


Subject(s)
Cell Transformation, Viral , Hepatitis B Surface Antigens/metabolism , Hepatitis B virus/physiology , Hepatitis B/virology , Viral Envelope Proteins/metabolism , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Carcinoma, Hepatocellular/etiology , Disease Management , Gene Expression Regulation, Viral , Genetic Variation , Genome, Viral , Hepatitis B/complications , Hepatitis B/diagnosis , Hepatitis B/drug therapy , Hepatitis B virus/ultrastructure , Host-Pathogen Interactions , Humans , Liver Neoplasms/etiology , Molecular Diagnostic Techniques , Virion
18.
Int J Mol Sci ; 22(12)2021 Jun 16.
Article in English | MEDLINE | ID: mdl-34208758

ABSTRACT

The goal of this study was to identify human papillomavirus (HPV) type 52 genetic and epigenetic changes associated with high-grade cervical precancer and cancer. Patients were selected from the HPV Persistence and Progression (PaP) cohort, a cervical cancer screening program at Kaiser Permanente Northern California (KPNC). We performed a nested case-control study of 89 HPV52-positive women, including 50 cases with predominantly cervical intraepithelial neoplasia grade 3 (CIN3) and 39 controls without evidence of abnormalities. We conducted methylation analyses using Illumina sequencing and viral whole genome Sanger sequencing. Of the 24 CpG sites examined, increased methylation at CpG site 5615 in HPV52 L1 region was the most significantly associated with CIN3, with a difference in median methylation of 17.9% (odds ratio (OR) = 4.8, 95% confidence interval (CI) = 1.9-11.8) and an area under the curve of 0.73 (AUC; 95% CI = 0.62-0.83). Complete genomic sequencing of HPV52 isolates revealed associations between SNPs present in sublineage C2 and a higher risk of CIN3, with ORs ranging from 2.8 to 3.3. This study identified genetic and epigenetic HPV52 variants associated with high risk for cervical precancer, improving the potential for early diagnosis of cervical neoplasia caused by HPV52.


Subject(s)
Alphapapillomavirus/genetics , Disease Susceptibility , Epigenesis, Genetic , Genetic Variation , Oncogene Proteins, Viral/genetics , Papillomavirus Infections/complications , Uterine Cervical Neoplasms/etiology , Alphapapillomavirus/classification , Cell Transformation, Viral , CpG Islands , DNA Methylation , Female , Genome, Viral , High-Throughput Nucleotide Sequencing , Humans , Papillomavirus Infections/virology , Phylogeny , Uterine Cervical Neoplasms/diagnosis
19.
Life Sci Alliance ; 4(9)2021 09.
Article in English | MEDLINE | ID: mdl-34290079

ABSTRACT

Chronic hepatitis B virus (HBV) infection is a major cause of hepatocellular carcinoma (HCC) world-wide. The molecular mechanisms of viral hepatocarcinogenesis are still partially understood. Here, we applied two complementary single-cell RNA-sequencing protocols to investigate HBV-HCC host cell interactions at the single cell level of patient-derived HCC. Computational analyses revealed a marked HCC heterogeneity with a robust and significant correlation between HBV reads and cancer cell differentiation. Viral reads significantly correlated with the expression of HBV-dependency factors such as HLF in different tumor compartments. Analyses of virus-induced host responses identified previously undiscovered pathways mediating viral carcinogenesis, such as E2F- and MYC targets as well as adipogenesis. Mapping of fused HBV-host cell transcripts allowed the characterization of integration sites in individual cancer cells. Collectively, single-cell RNA-Seq unravels heterogeneity and compartmentalization of both, virus and cancer identifying new candidate pathways for viral hepatocarcinogenesis. The perturbation of pro-carcinogenic gene expression even at low HBV levels highlights the need of HBV cure to eliminate HCC risk.


Subject(s)
Carcinoma, Hepatocellular/etiology , Cell Transformation, Viral , Hepatitis B virus/physiology , Hepatitis B/complications , Hepatitis B/virology , Liver Neoplasms/etiology , Adult , Aged , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Differentiation , Cell Line, Tumor , Disease Susceptibility , Female , Gene Expression Profiling , Gene Expression Regulation, Viral , Hepatitis B, Chronic/complications , Hepatitis B, Chronic/virology , Humans , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Grading , RNA, Viral , Single-Cell Analysis/methods , Transcriptome , Viral Load
20.
Virology ; 562: 1-8, 2021 10.
Article in English | MEDLINE | ID: mdl-34242747

ABSTRACT

Bovine leukemia virus (BLV) infection has spread worldwide causing significant economic losses in the livestock industry. In countries with a high prevalence of BLV, minimizing economic losses is challenging; thus, research into various countermeasures is important for improving BLV control. Because anti-BLV drugs have not been developed, the present study explored a promising chemical compound with anti-BLV activity. Initially, screening of a chemical compound library revealed that violaceoid E (vioE), which is isolated from fungus, showed antiviral activity. Further analysis demonstrated that the antiviral effect of vioE inhibited transcriptional activation of BLV. Cellular thermal shift assay and pulldown assays provided evidence for a direct interaction between vioE and the viral transactivator protein, Tax. These data indicate that interference with Tax-dependent transcription could be a novel target for development of anti-BLV drugs. Therefore, it is suggested that vioE is a novel antiviral compound against BLV.


Subject(s)
Antiviral Agents/pharmacology , Leukemia Virus, Bovine/drug effects , Animals , Antiviral Agents/chemistry , Cats , Cattle , Cell Line, Transformed , Cell Survival/drug effects , Cell Transformation, Viral/drug effects , Gene Products, tax/antagonists & inhibitors , Humans , Transcriptional Activation/drug effects , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...