Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 294
Filter
1.
Med ; 4(6): 347-352, 2023 Jun 09.
Article in English | MEDLINE | ID: mdl-37301195

ABSTRACT

The majority of oncogenic viruses are capable of integrating into the host genome, posing significant challenges to clinical control. Recent conceptual and technological advances, however, offer promising clinical applications. Here, we summarize the advances in our understanding of oncogenic viral integration, their clinical relevance, and the future perspectives.


Subject(s)
Genome , Oncogenic Viruses , Oncogenic Viruses/genetics , Virus Integration/genetics
2.
Int J Mol Sci ; 24(11)2023 May 31.
Article in English | MEDLINE | ID: mdl-37298494

ABSTRACT

Globally, viral infections substantially contribute to cancer development. Oncogenic viruses are taxonomically heterogeneous and drive cancers using diverse strategies, including epigenomic dysregulation. Here, we discuss how oncogenic viruses disrupt epigenetic homeostasis to drive cancer and focus on how virally mediated dysregulation of host and viral epigenomes impacts the hallmarks of cancer. To illustrate the relationship between epigenetics and viral life cycles, we describe how epigenetic changes facilitate the human papillomavirus (HPV) life cycle and how changes to this process can spur malignancy. We also highlight the clinical impact of virally mediated epigenetic changes on cancer diagnosis, prognosis, and treatment.


Subject(s)
Neoplasms , Viruses , Humans , Oncogenic Viruses/genetics , Epigenome , Neoplasms/pathology , Epigenesis, Genetic , DNA Methylation
3.
Int J Mol Sci ; 24(9)2023 Apr 25.
Article in English | MEDLINE | ID: mdl-37175509

ABSTRACT

Some viruses are known to be associated with the onset of specific cancers. These microorganisms, oncogenic viruses or oncoviruses, can convert normal cells into cancer cells by modulating the central metabolic pathways or hampering genomic integrity mechanisms, consequently inhibiting the apoptotic machinery and/or enhancing cell proliferation. Seven oncogenic viruses are known to promote tumorigenesis in humans: human papillomavirus (HPV), hepatitis B and C viruses (HBV, HCV), Epstein-Barr virus (EBV), human T-cell leukemia virus 1 (HTLV-1), Kaposi sarcoma-associated herpesvirus (KSHV), and Merkel cell polyomavirus (MCPyV). Recent research indicates that SARS-CoV-2 infection and COVID-19 progression may predispose recovered patients to cancer onset and accelerate cancer development. This hypothesis is based on the growing evidence regarding the ability of SARS-CoV-2 to modulate oncogenic pathways, promoting chronic low-grade inflammation and causing tissue damage. Herein, we summarize the main relationships known to date between virus infection and cancer, providing a summary of the proposed biochemical mechanisms behind the cellular transformation. Mechanistically, DNA viruses (such as HPV, HBV, EBV, and MCPyV) encode their virus oncogenes. In contrast, RNA viruses (like HCV, HTLV-1) may encode oncogenes or trigger host oncogenes through cis-/-trans activation leading to different types of cancer. As for SARS-CoV-2, its role as an oncogenic virus seems to occur through the inhibition of oncosuppressors or controlling the metabolic and autophagy pathways in the infected cells. However, these effects could be significant in particular scenarios like those linked to severe COVID-19 or long COVID. On the other hand, looking at the SARS-CoV-2─cancer relationship from an opposite perspective, oncolytic effects and anti-tumor immune response were triggered by SARS-CoV-2 infection in some cases. In summary, our work aims to recall comprehensive attention from the scientific community to elucidate the effects of SARS-CoV-2 and, more in general, ß-coronavirus infection on cancer susceptibility for cancer prevention or supporting therapeutic approaches.


Subject(s)
COVID-19 , Epstein-Barr Virus Infections , Hepatitis C , Neoplasms , Papillomavirus Infections , Humans , SARS-CoV-2 , Epstein-Barr Virus Infections/complications , Papillomavirus Infections/complications , Post-Acute COVID-19 Syndrome , Herpesvirus 4, Human , COVID-19/complications , Neoplasms/pathology , Oncogenic Viruses/genetics , Cell Transformation, Neoplastic , Hepatitis C/complications
4.
J Med Virol ; 95(1): e28324, 2023 01.
Article in English | MEDLINE | ID: mdl-36401345

ABSTRACT

Dynamic alteration of the epitranscriptome exerts regulatory effects on the lifecycle of oncogenic viruses in vitro. However, little is known about these effects in vivo because of the general lack of suitable animal infection models of these viruses. Using a model of rapid-onset Marek's disease lymphoma in chickens, we investigated changes in viral and host messenger RNA (mRNA) N6-methyladenosine (m6 A) modification during Marek's disease virus (MDV) infection in vivo. We found that the expression of major epitranscriptomic proteins varies among viral infection phases, reprogramming both the viral and the host epitranscriptomes. Specifically, the methyltransferase-like 3 (METTL3)/14 complex was suppressed during the lytic and reactivation phases of the MDV lifecycle, whereas its expression was increased during the latent phase and in MDV-induced tumors. METTL3/14 overexpression inhibits, whereas METTL3/14 knockdown enhances, MDV gene expression and replication. These findings reveal the dynamic features of the mRNA m6 A modification program during viral replication in vivo, especially in relation to key pathways involved in tumorigenesis.


Subject(s)
Marek Disease , Animals , Marek Disease/genetics , Oncogenic Viruses/genetics , Chickens , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
J Med Virol ; 95(1): e28254, 2023 01.
Article in English | MEDLINE | ID: mdl-36284485

ABSTRACT

Head and neck cancers are unique in so far that two major oncogenic viruses, Epstein Barr virus (EBV) and Human papillomavirus (HPV) infect adjacent anatomy and cause nasopharyngeal and oropharyngeal cancers, respectively. Dominant recognized carcinogens are alcohol and tobacco but some head and neck cancers have been found to have mixed carcinogens (including betel leaf, areca nuts, slaked lime, viruses, etc.) involved in their oncogenesis and conversely, groups of patients with unknown or less dominant carcinogens involved in their development. These cancers may have had viral involvement in the past but then lost most of their viral nucleic acids (be they DNA and/or RNA) below a detection threshold, thus rendering them virus-negative. Some of these virus-negative tumors appear to have mutagenic signatures associated with virus-positive cancers,  for example, from the APOBEC defense mechanism which is known to mutate viral nucleic acids as well as cause collateral damage to host DNA, with subsequent development of strongly viral prejudiced mutational signatures. These mechanisms are likely to be less efficient at oncogenesis than traditional EBV and HPV oncogenes directly driving mutagenesis, thus accounting for the smaller frequencies of these cancers found. More profound investigations of these unusual tumors are warranted to dissect out these mechanistic pathways.


Subject(s)
Epstein-Barr Virus Infections , Head and Neck Neoplasms , Nucleic Acids , Papillomavirus Infections , Humans , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/genetics , Papillomavirus Infections/complications , Head and Neck Neoplasms/genetics , Oncogenic Viruses/genetics , Carcinogenesis , Carcinogens , Papillomaviridae/genetics
6.
Nucleic Acids Res ; 50(20): 11799-11819, 2022 11 11.
Article in English | MEDLINE | ID: mdl-36350639

ABSTRACT

The oncogenic Epstein-Barr virus (EBV) evades the immune system but has an Achilles heel: its genome maintenance protein EBNA1. Indeed, EBNA1 is essential for viral genome maintenance but is also highly antigenic. Hence, EBV seemingly evolved a system in which the glycine-alanine repeat (GAr) of EBNA1 limits the translation of its own mRNA to the minimal level to ensure its essential function, thereby, at the same time, minimizing immune recognition. Therefore, defining intervention points at which to interfere with GAr-based inhibition of translation is an important step to trigger an immune response against EBV-carrying cancers. The host protein nucleolin (NCL) plays a critical role in this process via a direct interaction with G-quadruplexes (G4) formed in the GAr-encoding sequence of the viral EBNA1 mRNA. Here we show that the C-terminal arginine-glycine-rich (RGG) motif of NCL is crucial for its role in GAr-based inhibition of translation by mediating interaction of NCL with G4 of EBNA1 mRNA. We also show that this interaction depends on the type I arginine methyltransferase family, notably PRMT1 and PRMT3: drugs or small interfering RNA that target these enzymes prevent efficient binding of NCL on G4 of EBNA1 mRNA and relieve GAr-based inhibition of translation and of antigen presentation. Hence, this work defines type I arginine methyltransferases as therapeutic targets to interfere with EBNA1 and EBV immune evasion.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Tumor Virus Infections , Humans , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , Immune System/metabolism , Oncogenic Viruses/genetics , Oncogenic Viruses/metabolism , Protein-Arginine N-Methyltransferases , Repressor Proteins , RNA, Messenger/metabolism , Tumor Virus Infections/drug therapy , Tumor Virus Infections/metabolism
7.
J Med Virol ; 94(10): 4677-4688, 2022 10.
Article in English | MEDLINE | ID: mdl-35652186

ABSTRACT

Cancer is still ranked as a leading cause of death according to estimates from the World Health Organization (WHO) and the strong link between tumor viruses and human cancers have been proved for almost six decades. Cell-free DNA (cfDNA) has drawn enormous attention for its dynamic, instant, and noninvasive advantages as one popular type of cancer biomarker. cfDNAs are mainly released from apoptotic cells and exosomes released from cancer cells, including those infected with viruses. Although cfDNAs are present at low concentrations in peripheral blood, they can reflect tumor load with high sensitivity. Considering the relevance of the tumor viruses to the associated cancers, cfDNAs derived from viruses may serve as good biomarkers for the early screening, diagnosis, and treatment monitoring. In this review, we summarize the methods and newly developed analytic techniques for the detection of cfDNAs from different body fluids, and discuss the implications of cfDNAs derived from different tumor viruses in the detection and treatment monitoring of virus-associated cancers. A better understanding of cfDNAs derived from tumor viruses may help formulate novel antitumoral strategies to decrease the burden of cancers that attributed to viruses.


Subject(s)
Cell-Free Nucleic Acids , Neoplasms , Biomarkers, Tumor , Cell-Free Nucleic Acids/genetics , DNA, Neoplasm/genetics , Humans , Neoplasms/diagnosis , Neoplasms/pathology , Oncogenic Viruses/genetics
8.
Tumour Virus Res ; 14: 200239, 2022 12.
Article in English | MEDLINE | ID: mdl-35636683

ABSTRACT

Human tumor viruses cause various human cancers that account for at least 15% of the global cancer burden. Among the currently identified human tumor viruses, two are small DNA tumor viruses: human papillomaviruses (HPVs) and Merkel cell polyomavirus (MCPyV). The study of small DNA tumor viruses (adenoviruses, polyomaviruses, and papillomaviruses) has facilitated several significant biological discoveries and established some of the first animal models of virus-associated cancers. The development and use of preclinical in vivo models to study HPVs and MCPyV and their role in human cancer is the focus of this review. Important considerations in the design of animal models of small DNA tumor virus infection and disease, including host range, cell tropism, choice of virus isolates, and the ability to recapitulate human disease, are presented. The types of infection-based and transgenic model strategies that are used to study HPVs and MCPyV, including their strengths and limitations, are also discussed. An overview of the current models that exist to study HPV and MCPyV infection and neoplastic disease are highlighted. These comparative models provide valuable platforms to study various aspects of virus-associated human disease and will continue to expand knowledge of human tumor viruses and their relationship with their hosts.


Subject(s)
Merkel cell polyomavirus , Neoplasms , Polyomavirus Infections , Polyomavirus , Tumor Virus Infections , Animals , Humans , Merkel cell polyomavirus/genetics , Polyomavirus Infections/pathology , Tumor Virus Infections/complications , Neoplasms/genetics , Oncogenic Viruses/genetics
9.
Mol Ther ; 30(6): 2153-2162, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35143960

ABSTRACT

Cancer is a disease caused by loss of regulatory processes that control the cell cycle, resulting in increased proliferation. The loss of control can deregulate both tumor suppressors and oncogenes. Apart from cell intrinsic gene mutations and environmental factors, infection by cancer-causing viruses also induces changes that lead to malignant transformation. This can be caused by both expression of oncogenic viral proteins and also by changes in cellular genes and proteins that affect the epigenome. Thus, these epigenetic modifiers are good therapeutic targets, and several epigenetic inhibitors are approved for the treatment of different cancers. In addition to small molecule drugs, biological therapies, such as antibodies and viral therapies, are also increasingly being used to treat cancer. An HSV-1-derived oncolytic virus is currently approved by the US FDA and the European Medicines Agency. Similarly, an adenovirus-based therapeutic is approved for use in China for some cancer types. Because viruses can affect cellular epigenetics, the interaction of epigenome-targeting drugs with oncogenic and oncolytic viruses is a highly significant area of investigation. Here, we will review the current knowledge about the impact of using epigenetic drugs in tumors positive for oncogenic viruses or as therapeutic combinations with oncolytic viruses.


Subject(s)
Histones , Neoplasms , Oncogenic Viruses , Oncolytic Viruses , Histones/genetics , Humans , Neoplasms/genetics , Neoplasms/therapy , Oncogenic Viruses/genetics , Oncolytic Virotherapy , Oncolytic Viruses/genetics
10.
Anim Biotechnol ; 33(7): 1760-1765, 2022 Dec.
Article in English | MEDLINE | ID: mdl-33928832

ABSTRACT

Avian oncogenic or tumor diseases are common in poultry industry causing significant economic loss. Marek's disease (MD), avian leukosis (AL) and Reticuloendotheliosis (RE) are the three major viral oncogenic infections that are difficult to differentiate with gross lesions. Multiplex PCR for simultaneous detection and differentiation of these three viruses was developed and validated. The primers targeting the genes of pp38, pol and LTR for MDV, ALV and REV were designed to yield 206, 429, and 128 bp, respectively. The sensitivity of the PCR primers was checked with serial dilution of positive template DNA for each virus and found to be in the range of 10-5 to 10-7 of 1 µg/µl of initial template DNA. Out of 114 suspected tumor samples screened, 8 samples were positive for MDV, 13 samples were positive for ALV and 31 samples positive for REV. Five samples were positive for both MD and ALV; 3 samples were positive for MD and REV and 25 samples were positive for ALV and REV. Eight samples were positive for all three viruses. Multiplex PCR demonstrated to be a useful technique for simultaneous, rapid detection and differentiation of major tumor causing and immunosuppressive viral diseases of chicken.


Subject(s)
Marek Disease , Neoplasms , Poultry Diseases , Animals , Chickens/genetics , Multiplex Polymerase Chain Reaction/veterinary , Multiplex Polymerase Chain Reaction/methods , Oncogenic Viruses/genetics , Marek Disease/diagnosis , Marek Disease/pathology , Poultry Diseases/diagnosis , Poultry Diseases/pathology
11.
BMC Cancer ; 21(1): 1183, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34740324

ABSTRACT

BACKGROUND: Viral infections are prevalent in human cancers and they have great diagnostic and theranostic values in clinical practice. Recently, their potential of shaping the tumor immune microenvironment (TIME) has been related to the immunotherapy of human cancers. However, the landscape of viral expressions and immune status in human cancers remains incompletely understood. METHODS: We developed a next-generation sequencing (NGS)-based pipeline to detect viral sequences from the whole transcriptome and used machine learning algorithms to classify different TIME subtypes. RESULTS: We revealed a pan-cancer landscape of viral expressions in human cancers where 9 types of viruses were detected in 744 tumors of 25 cancer types. Viral infections showed different tissue tendencies and expression levels. Multi-omics analyses further revealed their distinct impacts on genomic, transcriptomic and immune responses. Epstein-Barr virus (EBV)-infected stomach adenocarcinoma (STAD) and Human Papillomavirus (HPV)-infected head and neck squamous cell carcinoma (HNSC) showed decreased genomic variations, significantly altered gene expressions, and effectively triggered anti-viral immune responses. We identified three TIME subtypes, in which the "Immune-Stimulation" subtype might be the promising candidate for immunotherapy. EBV-infected STAD and HPV-infected HNSC showed a higher frequency of the "Immune-Stimulation" subtype. Finally, we constructed the eVIIS pipeline to simultaneously evaluate viral infection and immune status in external datasets. CONCLUSIONS: Viral infections are prevalent in human cancers and have distinct influences on hosts. EBV and HPV infections combined with the TIME subtype could be promising biomarkers of immunotherapy in STAD and HNSC, respectively. The eVIIS pipeline could be a practical tool to facilitate clinical practice and relevant studies.


Subject(s)
Immunotherapy , Machine Learning , Neoplasms , Oncogenic Viruses , Tumor Microenvironment , Tumor Virus Infections , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , DNA, Viral/genetics , Epstein-Barr Virus Infections , Genetic Variation , Genome, Viral , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/therapy , Head and Neck Neoplasms/virology , Herpesvirus 4, Human/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Kaplan-Meier Estimate , Leukocytes/classification , Leukocytes/cytology , Mutation , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/virology , Oncogenic Viruses/genetics , Oncogenic Viruses/immunology , Papillomaviridae/genetics , Papillomavirus Infections , RNA-Seq , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/virology , Stomach Neoplasms/immunology , Stomach Neoplasms/therapy , Stomach Neoplasms/virology , Support Vector Machine , Transcriptome , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor Virus Infections/genetics , Tumor Virus Infections/immunology
12.
mBio ; 12(4): e0170621, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34425696

ABSTRACT

The methylation of RNA at the N6 position of adenosine (m6A) orchestrates multiple biological processes to control development, differentiation, and cell cycle, as well as various aspects of the virus life cycle. How the m6A RNA modification pathway is regulated to finely tune these processes remains poorly understood. Here, we discovered the m6A reader YTHDF2 as a caspase substrate via proteome-wide prediction, followed by in vitro and in vivo validations. We further demonstrated that cleavage-resistant YTHDF2 blocks, while cleavage-mimicking YTHDF2 fragments promote, the replication of a common human oncogenic virus, Epstein-Barr virus (EBV). Intriguingly, our study revealed a feedback regulation between YTHDF2 and caspase-8 via m6A modification of CASP8 mRNA and YTHDF2 cleavage during EBV replication. Further, we discovered that caspases cleave multiple components within the m6A RNA modification pathway to benefit EBV replication. Our study establishes that caspase disarming of the m6A RNA modification machinery fosters EBV replication. IMPORTANCE The discovery of an N6-methyladenosine (m6A) RNA modification pathway has fundamentally altered our understanding of the central dogma of molecular biology. This pathway is controlled by methyltransferases (writers), demethylases (erasers), and specific m6A binding proteins (readers). Emerging studies have linked the m6A RNA modification pathway to the life cycle of various viruses. However, very little is known regarding how this pathway is subverted to benefit viral replication. In this study, we established an unexpected linkage between cellular caspases and the m6A modification pathway, which is critical to drive the reactivation of a common tumor virus, Epstein-Barr virus (EBV).


Subject(s)
Adenosine/metabolism , Caspases/metabolism , Herpesvirus 4, Human/genetics , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Virus Replication/genetics , Adenosine/chemistry , Caspases/genetics , Cell Line , Epstein-Barr Virus Infections , Herpesvirus 4, Human/physiology , Humans , Methylation , Oncogenic Viruses/genetics , Oncogenic Viruses/physiology
13.
Curr Opin Immunol ; 72: 277-285, 2021 10.
Article in English | MEDLINE | ID: mdl-34364035

ABSTRACT

Oncoviruses are viruses that can cause tumors. Seven viruses are currently recognized as oncogenic in humans: Epstein Barr virus (EBV), Kaposi sarcoma-associated herpesvirus (KSHV, also known as HHV8), human papillomaviruses (HPVs), hepatitis B virus (HBV), hepatitis C virus (HCV), human T-lymphotropic virus-1 (HTLV-1), and Merkel cell polyomavirus (MCPyV). The clinical phenotypes resulting from infection with these oncoviruses range from asymptomatic infection to invasive cancers. Patients with inborn errors of immunity (IEI) are prone to the development of infectious diseases caused by a narrow or broad spectrum of pathogens, including oncoviruses in some cases. Studies of patients with IEI have deepened our understanding of the non-redundant mechanisms underlying the control of EBV, HHV8 and HPV infections. The human genetic factors conferring predisposition to oncogenic HBV, HCV, HTLV-1 and MCPyV manifestations remain elusive. We briefly review here what is currently known about the IEI conferring predisposition to severe infection with oncoviruses.


Subject(s)
Genetic Predisposition to Disease , Genetic Variation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunity/genetics , Oncogenic Viruses/immunology , Tumor Virus Infections/etiology , Autoimmunity , Biomarkers , Humans , Mutation , Oncogenic Viruses/classification , Oncogenic Viruses/genetics , Phenotype , Species Specificity
14.
Viruses ; 13(8)2021 07 30.
Article in English | MEDLINE | ID: mdl-34452367

ABSTRACT

The BK polyomavirus (BKPyV), a representative of the family Polyomaviridae, is widespread in the human population. While the virus does not cause significant clinical symptoms in immunocompetent individuals, it is activated in cases of immune deficiency, both pharmacological and pathological. Infection with the BKPyV is of particular importance in recipients of kidney transplants or HSC transplantation, in which it can lead to the loss of the transplanted kidney or to haemorrhagic cystitis, respectively. Four main genotypes of the virus are distinguished on the basis of molecular differentiation. The most common genotype worldwide is genotype I, with a frequency of about 80%, followed by genotype IV (about 15%), while genotypes II and III are isolated only sporadically. The distribution of the molecular variants of the virus is associated with the region of origin. BKPyV subtype Ia is most common in Africa, Ib-1 in Southeast Asia, and Ib-2 in Europe, while Ic is the most common variant in Northeast Asia. The development of molecular methods has enabled significant improvement not only in BKPyV diagnostics, but in monitoring the effectiveness of treatment as well. Amplification of viral DNA from urine by PCR (Polymerase Chain Reaction) and qPCR Quantitative Polymerase Chain Reaction) is a non-invasive method that can be used to confirm the presence of the genetic material of the virus and to determine the viral load. Sequencing techniques together with bioinformatics tools and databases can be used to determine variants of the virus, analyse their circulation in populations, identify relationships between them, and investigate the directions of evolution of the virus.


Subject(s)
BK Virus/genetics , BK Virus/pathogenicity , Genetic Variation , Genome, Viral , Polyomavirus Infections/diagnosis , Animals , BK Virus/classification , DNA, Viral/genetics , Genomics , Genotype , Immunocompromised Host , Kidney/virology , Kidney Transplantation/adverse effects , Mice , Oncogenic Viruses/genetics , Oncogenic Viruses/pathogenicity , Pathology, Molecular/methods , Polyomavirus Infections/virology , Transplant Recipients , Tumor Virus Infections/virology , Viral Load
15.
Adv Sci (Weinh) ; 8(9): 2004958, 2021 05.
Article in English | MEDLINE | ID: mdl-33977077

ABSTRACT

Approximately 15% of human cancers are estimated to be attributed to viruses. Virus sequences can be integrated into the host genome, leading to genomic instability and carcinogenesis. Here, a new deep convolutional neural network (CNN) model is developed with attention architecture, namely DeepVISP, for accurately predicting oncogenic virus integration sites (VISs) in the human genome. Using the curated benchmark integration data of three viruses, hepatitis B virus (HBV), human herpesvirus (HPV), and Epstein-Barr virus (EBV), DeepVISP achieves high accuracy and robust performance for all three viruses through automatically learning informative features and essential genomic positions only from the DNA sequences. In comparison, DeepVISP outperforms conventional machine learning methods by 8.43-34.33% measured by area under curve (AUC) value enhancement in three viruses. Moreover, DeepVISP can decode cis-regulatory factors that are potentially involved in virus integration and tumorigenesis, such as HOXB7, IKZF1, and LHX6. These findings are supported by multiple lines of evidence in literature. The clustering analysis of the informative motifs reveales that the representative k-mers in clusters could help guide virus recognition of the host genes. A user-friendly web server is developed for predicting putative oncogenic VISs in the human genome using DeepVISP.


Subject(s)
Deep Learning , Hepatitis B virus/genetics , Herpesvirus 1, Human/genetics , Herpesvirus 4, Human/genetics , Neoplasms/virology , Oncogenic Viruses/genetics , Virus Integration/genetics , Cluster Analysis , Genome, Human/genetics , Genomic Instability/genetics , Humans , Neoplasms/genetics , Reproducibility of Results
16.
J Med Virol ; 93(8): 5065-5075, 2021 08.
Article in English | MEDLINE | ID: mdl-33942339

ABSTRACT

Viral infection has been implicated in the pathogenesis of a plethora of human diseases. Although antiviral therapies effectively confront the viral spread and infection, how to completely eradicate the viral genome from infected cells remains a challenge. In this study, we demonstrated the reversible switching of primary cells between normal and malignant states by an oncogenic virus Kaposi's sarcoma-associated herpesvirus (KSHV) and CRISPR/Cas9-mediated targeting of a major viral latent protein. Primary cells can be transformed into malignant status by infection of KSHV, while elimination of the KSHV genome from latent KSHV-infected cells reverses KSHV-transformed primary cells back to a "normal state" by CRISPR/Cas-mediated knockout of viral major latent gene LANA. As a proof of concept, we demonstrated efficient elimination of KSHV episome in KSHV-associated primary effusion lymphoma cells resulting in the induction of apoptosis by liposome-encapsulated CRISPR/Cas9 ribonucleoprotein complexes (Lipo/Cas9-LANAsgRNA). Our work illustrates CRISPR/Cas as a promising technology for eliminating oncogenic viruses from persistently infected cells by taking advantage of the genetic differences between viral and cellular genomes. Compared to traditional antiviral therapy, our study offer an approach for antagonizing human oncogenic virus-related cancers by directly targeting as well as clearing viral genomes.


Subject(s)
Antigens, Viral/genetics , CRISPR-Cas Systems , Cell Transformation, Neoplastic/genetics , Herpesvirus 8, Human/genetics , Nuclear Proteins/genetics , Oncogenic Viruses/genetics , Animals , Antigens, Viral/metabolism , Apoptosis , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Cell Cycle , Cell Proliferation , Gene Knockout Techniques , Genome, Viral/genetics , Herpesvirus 8, Human/pathogenicity , Humans , Lymphoma, Primary Effusion/pathology , Mesenchymal Stem Cells , Nuclear Proteins/metabolism , Oncogenic Viruses/pathogenicity , RNA, Guide, Kinetoplastida/genetics , Rats , Virus Latency/genetics
17.
Front Immunol ; 12: 640918, 2021.
Article in English | MEDLINE | ID: mdl-33833760

ABSTRACT

Epstein Barr virus (EBV) is one of the most successful pathogens in humans with more than 95% of the human adult population persistently infected. EBV infects only humans and threatens these with its potent growth transforming ability that readily allows for immortalization of human B cells in culture. Accordingly, it is also found in around 1-2% of human tumors, primarily lymphomas and epithelial cell carcinomas. Fortunately, however, our immune system has learned to control this most transforming human tumor virus in most EBV carriers, and it requires modification of EBV associated lymphomagenesis and its immune control by either co-infections, such as malaria, Kaposi sarcoma associated herpesvirus (KSHV) and human immunodeficiency virus (HIV), or genetic predispositions for EBV positive tumors to emerge. Some of these can be modelled in humanized mice that, therefore, provide a valuable platform to test curative immunotherapies and prophylactic vaccines against these EBV associated pathologies.


Subject(s)
Cell Transformation, Viral/immunology , Coinfection , Epstein-Barr Virus Infections/immunology , Lymphoma/virology , Animals , Carcinogenesis/immunology , Cell Transformation, Viral/genetics , Coinfection/genetics , Coinfection/immunology , Coinfection/virology , Disease Models, Animal , Herpesvirus 4, Human , Humans , Lymphoma/genetics , Lymphoma/immunology , Mice , Oncogenic Viruses/genetics , Oncogenic Viruses/immunology
18.
Virol J ; 18(1): 18, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33441159

ABSTRACT

Viral oncogenic transformation of healthy cells into a malignant state is a well-established phenomenon but took decades from the discovery of tumor-associated viruses to their accepted and established roles in oncogenesis. Viruses cause ~ 15% of know cancers and represents a significant global health burden. Beyond simply causing cellular transformation into a malignant form, a number of these cancers are augmented by a subset of viral factors that significantly enhance the tumor phenotype and, in some cases, are locked in a state of oncogenic addiction, and substantial research has elucidated the mechanisms in these cancers providing a rationale for targeted inactivation of the viral components as a treatment strategy. In many of these virus-associated cancers, the prognosis remains extremely poor, and novel drug approaches are urgently needed. Unlike non-specific small-molecule drug screens or the broad-acting toxic effects of chemo- and radiation therapy, the age of designer nucleases permits a rational approach to inactivating disease-causing targets, allowing for permanent inactivation of viral elements to inhibit tumorigenesis with growing evidence to support their efficacy in this role. Although many challenges remain for the clinical application of designer nucleases towards viral oncogenes; the uniqueness and clear molecular mechanism of these targets, combined with the distinct advantages of specific and permanent inactivation by nucleases, argues for their development as next-generation treatments for this aggressive group of cancers.


Subject(s)
Carcinogenesis/drug effects , Neoplasms/drug therapy , Neoplasms/virology , Oncogenic Viruses/genetics , Animals , Cell Line , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Viral/drug effects , Cell Transformation, Viral/genetics , Humans , Mice , Oncogenes , Oncogenic Viruses/pathogenicity
19.
RNA Biol ; 18(5): 809-817, 2021 05.
Article in English | MEDLINE | ID: mdl-33499700

ABSTRACT

Oncogenic viruses are associated with approximately 15% of human cancers. In viral infections, microRNAs play an important role in host-pathogen interactions. miR-21 is a highly conserved non-coding RNA that not only regulates the development of oncogenic viral diseases, but also responds to the regulation of intracellular signal pathways. Oncogenic viruses, including HBV, HCV, HPV, and EBV, co-evolve with their hosts and cause persistent infections. The upregulation of host miR-21 manipulates key cellular pathways to evade host immune responses and then promote viral replication. Thus, a better understanding of the role of miR-21 in viral infections may help us to develop effective genetically-engineered oncolytic virus-based therapies against cancer.


Subject(s)
Host-Pathogen Interactions/genetics , MicroRNAs/physiology , Oncogenic Viruses/pathogenicity , Tumor Virus Infections/genetics , Animals , Humans , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/virology , Oncogenic Viruses/genetics , Oncogenic Viruses/immunology , Tumor Virus Infections/immunology , Tumor Virus Infections/pathology , Tumor Virus Infections/virology , Virus Replication/genetics
20.
Biochim Biophys Acta Rev Cancer ; 1875(1): 188476, 2021 01.
Article in English | MEDLINE | ID: mdl-33186643

ABSTRACT

BACKGROUND: It is well-known that certain cancers are caused by viruses. However, viral oncogenesis is complex and only a small fraction of the infected people develop cancer. Indeed, a number of environmental factors can contribute to virally infected cells developing cancer hallmarks, promoting tumorigenesis. SCOPE OF REVIEW: The hit-and-run theory proposes that viruses facilitate the accumulation of mutations and promote genomic instability until the virus becomes dispensable for tumour maintenance. Indeed, several studies have reported viral genome, episome and/or oncogene loss in tumour cells without losing malignant phenotype. MAJOR CONCLUSIONS: The current evidence supports the clear contribution of certain viruses to develop cancers. Importantly, the evidence supporting the sustained maintenance of malignancy after the loss of viral "presence" is sufficient to support the hit-and-run hypothesis of viral cancer development. Long-term tracking of vaccination outcome over the decades will test this theory. GENERAL SIGNIFICANCE: If the hit-and-run theory is true, viruses might cause more cancers than previously thought and will have implications in the prevention of many cancers through implementing vaccination programs.


Subject(s)
Cell Transformation, Neoplastic/genetics , Neoplasms/genetics , Oncogenes/genetics , Oncogenic Viruses/genetics , Disease Progression , Genomic Instability/genetics , Humans , Mutation/genetics , Neoplasms/pathology , Neoplasms/virology , Oncogenic Viruses/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...