Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 152
Filter
1.
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
2.
Viruses ; 13(8)2021 07 29.
Article in English | MEDLINE | ID: mdl-34452353

ABSTRACT

Insulin-like growth factor-1 (IGF-1) and the IGF-1 receptor (IGF-1R) belong to the insulin-like growth factor family, and IGF-1 activates intracellular signaling pathways by binding specifically to IGF-1R. The interaction between IGF-1 and IGF-1R transmits a signal through a number of intracellular substrates, including the insulin receptor substrate (IRS) and the Src homology collagen (Shc) proteins, which activate two major intracellular signaling pathways: the phosphatidylinositol 3-kinase (PI3K)/AKT and mitogen-activated protein kinase (MAPK) pathways, specifically the extracellular signal-regulated kinase (ERK) pathways. The PI3K/AKT kinase pathway regulates a variety of cellular processes, including cell proliferation and apoptosis. IGF1/IGF-1R signaling also promotes cell differentiation and proliferation via the Ras/MAPK pathway. Moreover, upon IGF-1R activation of the IRS and Shc adaptor proteins, Shc stimulates Raf through the GTPase Ras to activate the MAPKs ERK1 and ERK2, phosphorylate and several other proteins, and to stimulate cell proliferation. The IGF-1 signaling pathway is required for certain viral effects in oncogenic progression and may be induced as an effect of viral infection. The mechanisms of IGF signaling in animal viral infections need to be clarified, mainly because they are involved in multifactorial signaling pathways. The aim of this review is to summarize the current data obtained from virological studies and to increase our understanding of the complex role of the IGF-1 signaling axis in animal virus infections.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Signal Transduction/immunology , Virus Diseases/immunology , Virus Diseases/metabolism , Animals , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/immunology , Mice , Oncogenic Viruses/immunology , Oncogenic Viruses/metabolism , Phosphorylation , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Signal Transduction/genetics
3.
Viruses ; 12(7)2020 07 14.
Article in English | MEDLINE | ID: mdl-32674309

ABSTRACT

Basic leucine zipper (bZIP) transcription factors (TFs) govern diverse cellular processes and cell fate decisions. The hallmark of the leucine zipper domain is the heptad repeat, with leucine residues at every seventh position in the domain. These leucine residues enable homo- and heterodimerization between ZIP domain α-helices, generating coiled-coil structures that stabilize interactions between adjacent DNA-binding domains and target DNA substrates. Several cancer-causing viruses encode viral bZIP TFs, including human T-cell leukemia virus (HTLV), hepatitis C virus (HCV) and the herpesviruses Marek's disease virus (MDV), Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV). Here, we provide a comprehensive review of these viral bZIP TFs and their impact on viral replication, host cell responses and cell fate.


Subject(s)
Basic-Leucine Zipper Transcription Factors/physiology , Oncogenic Viruses/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Deltaretrovirus/genetics , Deltaretrovirus/metabolism , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , Humans , Mardivirus/genetics , Mardivirus/metabolism , Phylogeny , Tumor Virus Infections/metabolism , Tumor Virus Infections/virology , Unfolded Protein Response
4.
Biochim Biophys Acta Mol Basis Dis ; 1866(10): 165885, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32574835

ABSTRACT

Oncogenic viruses are among the apparent causes of cancer-associated mortality. It was estimated that 12% to 15% of human malignancies are linked to oncoviruses. Although modernist strategies and traditional genetic studies have defined host-pathogen interactions of the oncoviruses, their host functions which are critical for the establishment of infection still remain mysterious. However, over the last few years, it has become clear that infections hijack and modify cellular pathways for their benefit. In this context, we constructed the virus-host protein interaction networks of seven oncoviruses (EBV, HBV, HCV, HTLV-1, HHV8, HPV16, and HPV18), and revealed cellular pathways hijacking as a result of oncogenic virus infection. Several signaling pathways/processes such as TGF-ß signaling, cell cycle, retinoblastoma tumor suppressor protein, and androgen receptor signaling were mutually targeted by viruses to induce oncogenesis. Besides, cellular pathways specific to a certain virus were detected. By this study, we believe that we improve the understanding of the molecular pathogenesis of viral oncogenesis and provide information in setting new targets for treatment, prognosis, and diagnosis.


Subject(s)
Carcinogenesis/metabolism , Host-Pathogen Interactions , Neoplasms/metabolism , Oncogenic Viruses/pathogenicity , Protein Interaction Maps , Humans , Neoplasms/pathology , Neoplasms/virology , Oncogenic Viruses/metabolism , Protein Interaction Mapping , Signal Transduction , Viral Proteins/metabolism
5.
Proc Natl Acad Sci U S A ; 117(3): 1722-1730, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31919284

ABSTRACT

Viruses activate inflammasomes but then subvert resulting inflammatory responses to avoid elimination. We asked whether viruses could instead use such activated or primed inflammasomes to directly aid their propagation and spread. Since herpesviruses are experts at coopting cellular functions, we investigated whether Epstein-Barr virus (EBV), an oncoherpesvirus, exploits inflammasomes to activate its replicative or lytic phase. Indeed, our experiments reveal that EBV exploits several inflammasome sensors to actually activate its replicative phase from quiescence/latency. In particular, TXNIP, a key inflammasome intermediary, causes assembly of the NLRP3 inflammasome, resulting in caspase-1-mediated depletion of the heterochromatin-inducing epigenetic repressor KAP1/TRIM28 in a subpopulation of cells. As a result, only TXNIPhiKAP1lo cells, that is, in a primed/prolytic state, turn expression of the replication/lytic/reactivation switch protein on to enter the replicative phase. Our findings 1) demonstrate that EBV dovetails its escape strategy to a key cellular danger-sensing mechanism, 2) indicate that transcription may be regulated by KAP1 abundance aside from canonical regulation through its posttranslational modification, 3) mechanistically link diabetes, which frequently activates the NLRP3 inflammasome, to deregulation of a tumor virus, and 4) demonstrate that B lymphocytes from NOMID (neonatal onset multisystem inflammatory disease) patients who have NLRP3 mutations and suffer from hyperactive innate responses are defective in controlling a herpesvirus.


Subject(s)
Inflammasomes/metabolism , Inflammasomes/pharmacology , Oncogenic Viruses/drug effects , Oncogenic Viruses/metabolism , Virus Replication/drug effects , Virus Replication/physiology , B-Lymphocytes/metabolism , Carrier Proteins , Caspase 1/metabolism , Cell Line , Glucose/metabolism , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/metabolism , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Tripartite Motif-Containing Protein 28
6.
Discov Med ; 27(148): 163-166, 2019 03.
Article in English | MEDLINE | ID: mdl-31095925

ABSTRACT

Breast cancer is the most common form of cancer in women. The cause of sporadic cases is usually difficult to ascertain. Viruses that might be related to breast cancer are human papillomaviruses and herpes viruses. Mouse mammary tumor virus (MMTV) has also been a suspect. MMTV is a milk-transmitted beta retrovirus, a form of single-stranded positive-sense RNA virus that inserts a copy of its genome into the DNA of a host cell, thus altering the cell's genome. MMTV DNA sequences have been found in 36% of human breast tumor samples and 24% of non-cancerous breast tissue. The sequences were 98% similar to the MMTV envelope (env) gene. But a search for MMTV sequences within the human genome found no env sequences, although there were sequences from the MMTV GAGdUTPase and POL genes. Therefore, env sequences from breast tumors and normal breast tissue identified in other studies may have come from an MMTV infection. Humans apparently acquire MMTV infection from one species of mice, Mus domesticus. MMTV transmission from mice to humans may explain the relationship of breast cancer to high socioeconomic status. Many infectious diseases are associated with low income and poverty. The association is usually detrimental, but not always. During the 20th century, improved sanitation delayed exposure to the poliovirus and onset of infection in middle and upper class children. These children contracted paralytic polio, while children from low-income families living in poor neighborhoods and substandard housing, infected in infancy, were spared. Humoral immunity passively transferred from the mother protected them from paralytic polio, and they remained immune for life. A similar relationship may exist with MMTV. High income and affluence are linked to increasedbreastcancer incidence. Girls of high socioeconomic status living in affluent, clean homes would have delayed exposure to Mus domesticus and MMTV. When infection finally occurred they would be vulnerable to MMTV-induced breast cancer in later life. Impoverished girls living in substandard, mouse-infested housing would be exposed to mice and MMTV in early life. Humoral MMTV immunity passively transferred from the mother would protect them and render them immune to MMTV-induced breast cancer in later life.


Subject(s)
Breast Neoplasms , Mammary Neoplasms, Animal , Oncogenic Viruses/metabolism , Tumor Virus Infections , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/virology , Cattle , Female , Genome, Human , Humans , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/virology , Mice , Tumor Virus Infections/metabolism , Tumor Virus Infections/pathology , Tumor Virus Infections/virology
7.
Article in English | MEDLINE | ID: mdl-29254980

ABSTRACT

The term "neoantigen," as applied to molecules newly expressed on tumor cells, has a long history. The groundbreaking discovery of a cancer causing virus in chickens by Rous over 100 years ago, followed by discoveries of other tumor-causing viruses in animals, suggested a viral etiology of human cancers. The search for other oncogenic viruses in the 1960s and 1970s resulted in the discoveries of Epstein-Barr virus (EBV), hepatitis B virus (HBV), and human papilloma virus (HPV), and continues until the present time. Contemporaneously, the budding field of immunology was posing the question can the immune system of animals or humans recognize a tumor that develops from one's own tissues and what types of antigens would distinguish the tumor from normal cells. Molecules encoded by oncogenic viruses provided the most logical candidates and evidence was quickly gathered for both humoral and cellular recognition of viral antigens, referred to as neoantigens. Often, however, serologic responses to virus-bearing tumors revealed neoantigens unrelated to viral proteins and expressed on multiple tumor types, foreshadowing later findings of multiple changes in other genes in tumor cells creating nonviral neoantigens.


Subject(s)
Antigens, Viral/classification , Antigens, Viral/immunology , Cancer Vaccines/immunology , Neoplasms/therapy , Animals , Antigens, Viral/genetics , Humans , Neoplasms/prevention & control , Oncogenic Viruses/immunology , Oncogenic Viruses/metabolism
8.
Article in English | MEDLINE | ID: mdl-29254981

ABSTRACT

Recent work by several groups has undoubtedly shown that we can produce cancer vaccines targeting neoantigens. However, each vaccine is essentially a single-use, patient-specific product, making this approach resource-intensive. For this reason, it is important to ask whether this approach will be any more successful than what has been attempted during the last 30 years using vaccines targeting self-epitopes. Here, we discuss what might be expected from neoantigen vaccines based on our experience in chronic viral infections, and how this new approach may be applied to cancer immunotherapy.


Subject(s)
Antigens, Viral/immunology , Cancer Vaccines/immunology , Neoplasms/therapy , Oncogenic Viruses/immunology , Humans , Oncogenic Viruses/metabolism , T-Lymphocytes/physiology
9.
Mol Cell Proteomics ; 16(4 suppl 1): S65-S74, 2017 04.
Article in English | MEDLINE | ID: mdl-28104704

ABSTRACT

Oncogenic viruses are responsible for about 15% human cancers. This article explores the promise and challenges of viral proteomics in the study of the oncogenic human DNA viruses, HPV, McPyV, EBV and KSHV. These viruses have coevolved with their hosts and cause persistent infections. Each virus encodes oncoproteins that manipulate key cellular pathways to promote viral replication and evade the host immune response. Viral proteomics can identify cellular pathways perturbed by viral infection, identify cellular proteins that are crucial for viral persistence and oncogenesis, and identify important diagnostic and therapeutic targets.


Subject(s)
Neoplasms/virology , Oncogene Proteins/metabolism , Oncogenic Viruses/physiology , Proteomics/methods , DNA Virus Infections/immunology , DNA Virus Infections/virology , Host-Pathogen Interactions , Humans , Oncogenic Viruses/metabolism , Viral Proteins/metabolism , Virus Replication
10.
Cold Spring Harb Protoc ; 2014(11): 1128-35, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-25368315

ABSTRACT

For successful infection, avian sarcoma leukosis virus subgroup A (ASLV-A) requires its receptor, tumor virus A (TVA), to be present on the surface of target cells. This is the basis of the RCAS-TVA gene delivery system: Mammalian cells lack the gene encoding TVA and are normally resistant to infection by ASLV; however, transgenic targeting of TVA to specific cell types or tissues in the mouse renders these cells uniquely susceptible to infection by ASLV-A-based RCAS viruses. The RCAS-TVA system is a powerful tool for effectively modeling human tumors, including pancreatic, ovarian, and breast cancers, gliomas, and melanomas. RCAS viruses can deliver cDNAs (≤2.8 kb), as well as short hairpin RNAs (shRNAs), microRNAs (miRNAs), and other noncoding RNAs. Compared with traditional transgenic and knockout mice, the RCAS-TVA system has several strengths. First, virus delivery is generally performed postnatally and results in a relatively low infection rate of target cells; the sporadic postnatal expression of the gene of interest mimics the situation in developing human tumors. Second, a single transgenic mouse line can be used to compare the consequences of specific genes on tumor development, with viruses encoding oncogenes or shRNAs targeting specific tumor suppressor genes. TVA mouse strains can also be easily combined with transgenic, knock-in, and knockout mouse models to study cooperating genetic events.


Subject(s)
Avian Sarcoma Viruses/genetics , Disease Models, Animal , Gene Transfer Techniques , Genetic Vectors , Neoplasms/pathology , Oncogenic Viruses/genetics , Receptors, Virus/metabolism , Animals , Humans , Mice , Oncogenic Viruses/metabolism , Receptors, Virus/genetics
11.
Nature ; 487(7408): 491-5, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22810586

ABSTRACT

Genotypic differences greatly influence susceptibility and resistance to disease. Understanding genotype-phenotype relationships requires that phenotypes be viewed as manifestations of network properties, rather than simply as the result of individual genomic variations. Genome sequencing efforts have identified numerous germline mutations, and large numbers of somatic genomic alterations, associated with a predisposition to cancer. However, it remains difficult to distinguish background, or 'passenger', cancer mutations from causal, or 'driver', mutations in these data sets. Human viruses intrinsically depend on their host cell during the course of infection and can elicit pathological phenotypes similar to those arising from mutations. Here we test the hypothesis that genomic variations and tumour viruses may cause cancer through related mechanisms, by systematically examining host interactome and transcriptome network perturbations caused by DNA tumour virus proteins. The resulting integrated viral perturbation data reflects rewiring of the host cell networks, and highlights pathways, such as Notch signalling and apoptosis, that go awry in cancer. We show that systematic analyses of host targets of viral proteins can identify cancer genes with a success rate on a par with their identification through functional genomics and large-scale cataloguing of tumour mutations. Together, these complementary approaches increase the specificity of cancer gene identification. Combining systems-level studies of pathogen-encoded gene products with genomic approaches will facilitate the prioritization of cancer-causing driver genes to advance the understanding of the genetic basis of human cancer.


Subject(s)
Genes, Neoplasm/genetics , Genome, Human/genetics , Host-Pathogen Interactions , Neoplasms/genetics , Neoplasms/metabolism , Oncogenic Viruses/pathogenicity , Viral Proteins/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Adenoviridae/pathogenicity , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions/genetics , Humans , Neoplasms/pathology , Oncogenic Viruses/genetics , Oncogenic Viruses/metabolism , Open Reading Frames/genetics , Papillomaviridae/genetics , Papillomaviridae/metabolism , Papillomaviridae/pathogenicity , Polyomavirus/genetics , Polyomavirus/metabolism , Polyomavirus/pathogenicity , Receptors, Notch/metabolism , Signal Transduction , Two-Hybrid System Techniques , Viral Proteins/genetics
12.
Mol Cancer Ther ; 11(1): 14-23, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22234809

ABSTRACT

Oncogenic viruses represent a significant public health burden in light of the multitude of malignancies that result from chronic or spontaneous viral infection and transformation. Although many of the molecular signaling pathways that underlie virus-mediated cellular transformation are known, the impact of these viruses on metabolic signaling and phenotype within proliferating tumor cells is less well understood. Whether the interaction of oncogenic viruses with metabolic signaling pathways involves enhanced glucose uptake and glycolysis (both hallmark features of transformed cells) or dysregulation of molecular pathways that regulate oxidative stress, viruses are adept at facilitating tumor expansion. Through their effects on cell proliferation pathways, such as the PI3K and MAPK pathways, the cell cycle regulatory proteins p53 and ATM, and the cell stress response proteins HIF-1α and AMPK, viruses exert control over critical metabolic signaling cascades. Additionally, oncogenic viruses modulate the tumor metabolomic profile through direct and indirect interactions with glucose transporters, such as GLUT1, and specific glycolytic enzymes, including pyruvate kinase, glucose 6-phosphate dehydrogenase, and hexokinase. Through these pathways, oncogenic viruses alter the phenotypic characteristics and energy-use methods of transformed cells; therefore, it may be possible to develop novel antiglycolytic therapies to target these dysregulated pathways in virus-derived malignancies.


Subject(s)
Glucose/metabolism , Metabolic Networks and Pathways , Neoplasms/virology , Oncogenic Viruses/metabolism , Cell Cycle , Cell Proliferation , Glycolysis , Humans , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Stress, Physiological
13.
FEBS Lett ; 585(20): 3174-84, 2011 Oct 20.
Article in English | MEDLINE | ID: mdl-21846466

ABSTRACT

Evidence over the last two decades from a number of disciplines has solidified some fundamental concepts in metastasis, a major contributor to cancer associated deaths. However, significant advances have been made in controlling this critical cellular process by focusing on targeted therapy. A key set of factors associated with this invasive phenotype is the nm23 family of over twenty metastasis-associated genes. Among the eight known isoforms, Nm23-H1 is the most studied potential anti-metastatic factor associated with human cancers. Importantly, a growing body of work has clearly suggested a critical role for Nm23-H1 in limiting tumor cell motility and progression induced by several tumor viruses, including Epstein-Barr virus (EBV), Kaposi's sarcoma associated herpes virus (KSHV) and human papilloma virus (HPV). A more in depth understanding of the interactions between tumor viruses encoded antigens and Nm23-H1 will facilitate the elucidation of underlying mechanism(s) which contribute to virus-associated cancers. Here, we review recent studies to explore the molecular links between human oncogenic viruses and progression of metastasis, in particular the deregulation of Nm23-H1 mediated suppression.


Subject(s)
Antigens, Neoplasm/metabolism , Antigens, Viral/metabolism , NM23 Nucleoside Diphosphate Kinases/metabolism , Oncogenic Viruses/metabolism , Tumor Suppressor Proteins/metabolism , Tumor Virus Infections/enzymology , Animals , Antigens, Neoplasm/genetics , Antigens, Viral/genetics , Cell Movement , Humans , NM23 Nucleoside Diphosphate Kinases/genetics , Neoplasm Metastasis , Oncogenic Viruses/genetics , Tumor Suppressor Proteins/genetics , Tumor Virus Infections/genetics , Tumor Virus Infections/pathology
14.
Cancer Lett ; 305(2): 200-17, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-20813452

ABSTRACT

It is well documented that viral genomes either inserted into the cellular DNA or co-replicating with it in episomal form can be lost from neoplastic cells. Therefore, "hit and run"-mechanisms have been a topic of longstanding interest in tumor virology. The basic idea is that the transient acquisition of a complete or incomplete viral genome may be sufficient to induce malignant conversion of host cells in vivo, resulting in neoplastic development. After eliciting a heritable change in the gene expression pattern of the host cell (initiation), the genomes of tumor viruses may be completely lost, i.e. in a hit and run-scenario they are not necessary for the maintenance of the malignant state. The expression of viral oncoproteins and RNAs may interfere not only with regulators of cell proliferation, but also with DNA repair mechanisms. DNA recombinogenic activities induced by tumor viruses or activated by other mechanisms may contribute to the secondary loss of viral genomes from neoplastic cells. Viral oncoproteins can also cause epigenetic dysregulation, thereby reprogramming cellular gene expression in a heritable manner. Thus, we expect that epigenetic scenarios of viral hit and run-tumorigenesis may facilitate new, innovative experiments and clinical studies in spite of the fact that the regular presence of a suspected human tumor virus in an early phase of neoplastic development and its subsequent regular loss have not been demonstrated yet. We propose that virus-specific "epigenetic signatures", i.e. alterations of the host cell epigenome, especially altered DNA methylation patterns, may help to identify viral hit and run-oncogenic events, even after the complete loss of tumor viruses from neoplastic cells.


Subject(s)
Epigenesis, Genetic , Animals , Cell Transformation, Neoplastic , Chromatin/metabolism , DNA Methylation , DNA Repair , Genes, Tumor Suppressor , Herpesvirus 4, Human/metabolism , Humans , Mice , Models, Genetic , Mutation , Oncogenic Viruses/metabolism , Recombination, Genetic , VDJ Recombinases/metabolism
15.
Cancer Lett ; 305(2): 186-99, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-20943311

ABSTRACT

Tumor viruses are a class of pathogens with well established roles in the development of malignant diseases. Numerous bodies of work have highlighted miRNAs (microRNAs) as critical regulators of tumor pathways and it is clear that the dysregulation of cellular miRNA expression can promote tumor formation. Tumor viruses encode their own miRNAs and/or manipulate the expression of cellular miRNAs to modulate their host cell environment, thereby facilitating their respective infection cycles. The modulation of these miRNA responsive pathways, however, often influences certain signal transduction cascades in ways that favor tumorigenesis. In this review, we discuss the roles of virally-encoded and virally-regulated cellular miRNAs in the respective viral life cycles and in virus associated pathogenesis.


Subject(s)
MicroRNAs/metabolism , Oncogenic Viruses/metabolism , Virus Diseases/metabolism , Animals , Gene Silencing , Herpesvirus 4, Human/metabolism , Herpesvirus 8, Human/metabolism , Humans , Models, Biological , Neoplasms/virology , Signal Transduction
16.
Cancer Lett ; 305(2): 218-27, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-20971551

ABSTRACT

The study of cancer is incomplete without taking into consideration of tumorigenic viruses. Initially, searches for human cancer viruses were fruitless despite an expansion of our knowledge in the same period concerning acute-transforming retroviruses in animals. However, over the last 40 years, we have witnessed rapid progress in the tumor virology field. Currently, acknowledged human cancer viruses include Epstein-Barr virus, hepatitis B virus, hepatitis C virus, high-risk human papilloma viruses, human T-cell lymphotropic virus type 1 and Kaposi's sarcoma-associated herpesvirus. Extensive epidemiological and mechanistic studies have led to the development of novel preventive and therapeutic approaches for managing some of these infections and associated cancers. In addition, recent advances in molecular technologies have enabled the discovery of a new potential human tumor virus, Merkel cell polyomavirus, but its association with cancer remains to be validated. It is anticipated that in the next few decades many additional human cancer viruses will be discovered and the mechanisms underlying viral oncogenesis delineated. Thus, it can be expected that better tools for preventing and treating virus-associated cancer will be available in the near future.


Subject(s)
Neoplasms/virology , Virus Diseases/virology , Viruses/metabolism , Animals , Cancer Vaccines , Endogenous Retroviruses/genetics , Hepacivirus/metabolism , Hepatitis B virus/metabolism , Herpesvirus 4, Human/metabolism , Herpesvirus 8, Human/metabolism , Human T-lymphotropic virus 1/metabolism , Humans , Neoplasms/complications , Oncogenic Viruses/metabolism , Papillomaviridae/metabolism , Polyomavirus/metabolism , Virus Diseases/complications
20.
J Natl Cancer Inst ; 101(7): 488-97, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19318640

ABSTRACT

BACKGROUND: The mechanism by which human cytomegalovirus (HCMV) stimulates oncogenesis is unclear. Because cellular immortalization and transformation require telomerase activation by expression of the telomerase reverse transcriptase (hTERT) gene, we examined the role of HCMV in telomerase activation. METHODS: Normal human diploid fibroblasts (HDFs) and human malignant glioma (MG) cell lines were infected with HCMV or transfected with expression vectors encoding HCMV immediate early (IE) antigen 72 or 86. hTERT expression and promoter activity and telomerase activity were evaluated using reverse transcription-polymerase chain reaction, a luciferase reporter assay, and a telomeric repeat amplification protocol, respectively. hTERT promoter occupancy by the transcription factor Sp1, IE antigens, and histone deacetylases (HDACs) was assessed by chromatin immunoprecipitation. hTERT and IE protein expression in human primary glioblastoma multiforme (GBM) was determined immunohistochemically. All statistical tests were two-sided. RESULTS: In telomerase and hTERT-negative HDFs, HCMV infection induced constitutive hTERT expression and telomerase activation. The hTERT promoter activity in HDFs and MG cell lines was statistically significantly enhanced by HCMV in a dose-dependent manner (mean luciferase activity [arbitrary units] in control HDFs and in HDFs infected with HCMV at multiplicities of infection [MOIs] of 0.1 = 6 and 521, respectively, difference = 515, 95% CI = 178 to 850; mean activity at MOI of 1 and 10 = 8828 and 59,923, respectively; P < .001 comparing control with HCMV-infected cells at all MOIs). Ectopic expression of HCMV IE-72 protein also stimulated hTERT promoter activity in HDFs. HCMV-mediated transactivation of the hTERT gene was dependent on the presence of Sp1-binding sites in the hTERT promoter and was accompanied by increases in Sp1 binding, acetylation of histone H3, and a reduction in HDAC binding at the core promoter. In specimens of GBM, HCMV IE and hTERT proteins were colocalized in malignant cells and their levels paralleled each other. CONCLUSIONS: HCMV activates telomerase in both HDFs and malignant cells. These findings begin to reveal a novel mechanism by which HCMV infection may be linked to or modulate oncogenesis through telomerase activation.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus/metabolism , Fibroblasts/metabolism , Glioma/enzymology , Telomerase/metabolism , Telomere/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Cytomegalovirus Infections/enzymology , Enzyme Activation , Fibroblasts/enzymology , Fluorescent Antibody Technique , Glioblastoma/metabolism , Glioma/metabolism , Glioma/virology , Humans , Immunoblotting , Immunohistochemistry , Luciferases/metabolism , Oncogenic Viruses/metabolism , RNA, Messenger/metabolism , Research Design , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/genetics , Telomere/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...