Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.624
Filter
1.
Glia ; 69(9): 2059-2076, 2021 09.
Article in English | MEDLINE | ID: mdl-33638562

ABSTRACT

Gliomas are the most common primary intrinsic brain tumors occurring in adults. Of all malignant gliomas, glioblastoma (GBM) is considered the deadliest tumor type due to diffuse brain invasion, immune evasion, cellular, and molecular heterogeneity, and resistance to treatments resulting in high rates of recurrence. An extensive understanding of the genomic and microenvironmental landscape of gliomas gathered over the past decade has renewed interest in pursuing novel therapeutics, including immune checkpoint inhibitors, glioma-associated macrophage/microglia (GAMs) modulators, and others. In light of this, predictive animal models that closely recreate the conditions and findings found in human gliomas will serve an increasingly important role in identifying new, effective therapeutic strategies. Although numerous syngeneic, xenograft, and transgenic rodent models have been developed, few include the full complement of pathobiological features found in human tumors, and therefore few accurately predict bench-to-bedside success. This review provides an update on how genetically engineered rodent models based on the replication-competent avian-like sarcoma (RCAS) virus/tumor virus receptor-A (tv-a) system have been used to recapitulate key elements of human gliomas in an immunologically intact host microenvironment and highlights new approaches using this model system as a predictive tool for advancing translational glioma research.


Subject(s)
Brain Neoplasms , Disease Models, Animal , Glioma , Sarcoma , Animals , Avian Sarcoma Viruses/genetics , Brain Neoplasms/pathology , Glioma/pathology , Humans , Oncogenic Viruses , Receptors, Virus , Tumor Microenvironment
2.
J Virol ; 94(12)2020 06 01.
Article in English | MEDLINE | ID: mdl-32238588

ABSTRACT

Tetherin/BST-2 is an antiviral protein that blocks the release of enveloped viral particles by linking them to the membrane of producing cells. At first, BST-2 genes were described only in humans and other mammals. Recent work identified BST-2 orthologs in nonmammalian vertebrates, including birds. Here, we identify the BST-2 sequence in domestic chicken (Gallus gallus) for the first time and demonstrate its activity against avian sarcoma and leukosis virus (ASLV). We generated a BST-2 knockout in chicken cells and showed that BST-2 is a major determinant of an interferon-induced block of ASLV release. Ectopic expression of chicken BST-2 blocks the release of ASLV in chicken cells and of human immunodeficiency virus type 1 (HIV-1) in human cells. Using metabolic labeling and pulse-chase analysis of HIV-1 Gag proteins, we verified that chicken BST-2 blocks the virus at the release stage. Furthermore, we describe BST-2 orthologs in multiple avian species from 12 avian orders. Previously, some of these species were reported to lack BST-2, highlighting the difficulty of identifying sequences of this extremely variable gene. We analyzed BST-2 genes in the avian orders Galliformes and Passeriformes and showed that they evolve under positive selection. This indicates that avian BST-2 is involved in host-virus evolutionary arms races and suggests that BST-2 antagonists exist in some avian viruses. In summary, we show that chicken BST-2 has the potential to act as a restriction factor against ASLV. Characterizing the interaction of avian BST-2 with avian viruses is important in understanding innate antiviral defenses in birds.IMPORTANCE Birds are important hosts of viruses that have the potential to cause zoonotic infections in humans. However, only a few antiviral genes (called viral restriction factors) have been described in birds, mostly because birds lack counterparts of highly studied mammalian restriction factors. Tetherin/BST-2 is a restriction factor, originally described in humans, that blocks the release of newly formed virus particles from infected cells. Recent work identified BST-2 in nonmammalian vertebrate species, including birds. Here, we report the BST-2 sequence in domestic chicken and describe its antiviral activity against a prototypical avian retrovirus, avian sarcoma and leukosis virus (ASLV). We also identify BST-2 genes in multiple avian species and show that they evolve rapidly in birds, which is an important indication of their relevance for antiviral defense. Analysis of avian BST-2 genes will shed light on defense mechanisms against avian viral pathogens.


Subject(s)
Avian Proteins/immunology , Avian Sarcoma Viruses/immunology , Bone Marrow Stromal Antigen 2/immunology , Evolution, Molecular , Galliformes/immunology , Sarcoma, Avian/immunology , Amino Acid Sequence , Animals , Avian Proteins/genetics , Avian Sarcoma Viruses/genetics , Avian Sarcoma Viruses/pathogenicity , Bone Marrow Stromal Antigen 2/genetics , Cell Line , Fibroblasts/immunology , Fibroblasts/virology , Galliformes/genetics , Galliformes/virology , Gene Expression Regulation , HEK293 Cells , HIV-1/genetics , HIV-1/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Passeriformes/genetics , Passeriformes/immunology , Passeriformes/virology , Sarcoma, Avian/genetics , Sarcoma, Avian/virology , Selection, Genetic , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Virus Release , Virus Replication , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology
3.
Viruses ; 11(6)2019 06 05.
Article in English | MEDLINE | ID: mdl-31195660

ABSTRACT

The subgroup A through E avian sarcoma and leukosis viruses (ASLV(A) through ASLV(E)) are a group of highly related alpharetroviruses that have evolved their envelope glycoproteins to use different receptors to enable efficient virus entry due to host resistance and/or to expand host range. Previously, we demonstrated that ASLV(A) in the presence of a competitor to the subgroup A Tva receptor, SUA-rIgG immunoadhesin, evolved to use other receptor options. The selected mutant virus, RCASBP(A)Δ155-160, modestly expanded its use of the Tvb and Tvc receptors and possibly other cell surface proteins while maintaining the binding affinity to Tva. In this study, we further evolved the Δ155-160 virus with the genetic selection pressure of a soluble form of the Tva receptor that should force the loss of Tva binding affinity in the presence of the Δ155-160 mutation. Viable ASLVs were selected that acquired additional mutations in the Δ155-160 Env hypervariable regions that significantly broadened receptor usage to include Tvb and Tvc as well as retaining the use of Tva as a receptor determined by receptor interference assays. A similar deletion in the hr1 hypervariable region of the subgroup C ASLV glycoproteins evolved to broaden receptor usage when selected on Tvc-negative cells.


Subject(s)
Avian Sarcoma Viruses/genetics , Receptors, Virus/physiology , Viral Envelope Proteins/genetics , Animals , Avian Proteins/physiology , Binding Sites/physiology , Biological Evolution , Cell Line , Chickens/virology , Glycoproteins/genetics , Mutation , Sarcoma, Avian/virology , Virus Internalization
4.
Viruses ; 11(6)2019 05 30.
Article in English | MEDLINE | ID: mdl-31151254

ABSTRACT

The initial step of retrovirus entry-the interaction between the virus envelope glycoprotein trimer and a cellular receptor-is complex, involving multiple, noncontiguous determinants in both proteins that specify receptor choice, binding affinity and the ability to trigger conformational changes in the viral glycoproteins. Despite the complexity of this interaction, retroviruses have the ability to evolve the structure of their envelope glycoproteins to use a different cellular protein as receptors. The highly homologous subgroup A to E Avian Sarcoma and Leukosis Virus (ASLV) glycoproteins belong to the group of class 1 viral fusion proteins with a two-step triggering mechanism that allows experimental access to intermediate structures during the fusion process. We and others have taken advantage of replication-competent ASLVs and exploited genetic selection strategies to force the ASLVs to naturally evolve and acquire envelope glycoprotein mutations to escape the pressure on virus entry and still yield a functional replicating virus. This approach allows for the simultaneous selection of multiple mutations in multiple functional domains of the envelope glycoprotein that may be required to yield a functional virus. Here, we review the ASLV family and experimental system and the reverse engineering approaches used to understand the evolution of ASLV receptor usage.


Subject(s)
Avian Leukosis Virus/genetics , Avian Sarcoma Viruses/genetics , Evolution, Molecular , Receptors, Virus/genetics , Reverse Genetics , Animals , Avian Sarcoma Viruses/classification , Chickens/virology , Mutation , Sarcoma, Avian , Viral Envelope Proteins/genetics , Virus Internalization , Virus Replication
5.
Viruses ; 11(6)2019 05 31.
Article in English | MEDLINE | ID: mdl-31159208

ABSTRACT

The subgroup A through E avian sarcoma and leukosis viruses ASLV(A) through ASLV(E) are a group of highly related alpharetroviruses that have evolved to use very different host protein families as receptors. We have exploited genetic selection strategies to force the replication-competent ASLVs to naturally evolve and acquire mutations to escape the pressure on virus entry and yield a functional replicating virus. In this study, evolutionary pressure was exerted on ASLV(B) virus entry and replication using a secreted for of its Tvb receptor. As expected, mutations in the ASLV(B) surface glycoprotein hypervariable regions were selected that knocked out the ability for the mutant glycoprotein to bind the sTvbS3-IgG inhibitor. However, the subgroup B Rous associated virus 2 (RAV-2) also required additional mutations in the C-terminal end of the SU glycoprotein and multiple regions of TM highlighting the importance of the entire viral envelope glycoprotein trimer structure to mediate the entry process efficiently. These mutations altered the normal two-step ASLV membrane fusion process to enable infection.


Subject(s)
Avian Leukosis Virus/genetics , Avian Sarcoma Viruses/genetics , Mutation , Receptors, Virus/genetics , Receptors, Virus/metabolism , Viral Envelope Proteins , Animals , Avian Leukosis Virus/physiology , Avian Sarcoma Viruses/physiology , Cell Line , Chick Embryo , Chickens/virology , Viral Envelope Proteins/genetics , Virus Replication
6.
Viruses ; 11(1)2019 01 18.
Article in English | MEDLINE | ID: mdl-30669277

ABSTRACT

Jan Svoboda triggered investigations on non-defective avian sarcoma viruses. These viruses were a critical factor in the genetic understanding of retroviruses. They provided the single and unique access to the field and facilitated the discovery of the first oncogene src and of the cellular origin of retroviral oncogenes. They continue to be of importance as singularly effective expression vectors that have provided insights into the molecular functions of numerous oncogenes. Combined with the contributions to the validation of the provirus hypothesis, Jan Svoboda's investigations of non-defective avian sarcoma viruses have shaped a large and important part of retrovirology.


Subject(s)
Avian Sarcoma Viruses/genetics , Genes, Viral , Oncogenes , Animals , Humans , Proviruses/genetics
7.
PLoS One ; 12(3): e0174557, 2017.
Article in English | MEDLINE | ID: mdl-28358926

ABSTRACT

Previously rodent preclinical research in gliomas frequently involved implantation of cell lines such as C6 and 9L into the rat brain. More recently, mouse models have taken over, the genetic manipulability of the mouse allowing the creation of genetically accurate models outweighed the disadvantage of its smaller brain size that limited time allowed for tumor progression. Here we illustrate a method that allows glioma formation in the rat using the replication competent avian-like sarcoma (RCAS) virus / tumor virus receptor-A (tv-a) transgenic system of post-natal cell type-specific gene transfer. The RCAS/tv-a model has emerged as a particularly versatile and accurate modeling technology by enabling spatial, temporal, and cell type-specific control of individual gene transformations and providing de novo formed glial tumors with distinct molecular subtypes mirroring human GBM. Nestin promoter-driven tv-a (Ntv-a) transgenic Sprague-Dawley rat founder lines were created and RCAS PDGFA and p53 shRNA constructs were used to initiate intracranial brain tumor formation. Tumor formation and progression were confirmed and visualized by magnetic resonance imaging (MRI) and spectroscopy. The tumors were analyzed using histopathological and immunofluorescent techniques. All experimental animals developed large, heterogeneous brain tumors that closely resembled human GBM. Median survival was 92 days from tumor initiation and 62 days from the first point of tumor visualization on MRI. Each tumor-bearing animal showed time dependent evidence of malignant progression to high-grade glioma by MRI and neurological examination. Post-mortem tumor analysis demonstrated the presence of several key characteristics of human GBM, including high levels of tumor cell proliferation, pseudopalisading necrosis, microvascular proliferation, invasion of tumor cells into surrounding tissues, peri-tumoral reactive astrogliosis, lymphocyte infiltration, presence of numerous tumor-associated microglia- and bone marrow-derived macrophages, and the formation of stem-like cell niches within the tumor. This transgenic rat model may enable detailed interspecies comparisons of fundamental cancer pathways and clinically relevant experimental imaging procedures and interventions that are limited by the smaller size of the mouse brain.


Subject(s)
Brain/diagnostic imaging , Glioma/genetics , Nestin/genetics , Platelet-Derived Growth Factor/genetics , Tumor Suppressor Protein p53/genetics , Animals , Avian Sarcoma Viruses/genetics , Avian Sarcoma Viruses/pathogenicity , Brain/pathology , Brain/virology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Genetic Engineering , Glioma/diagnostic imaging , Glioma/pathology , Glioma/virology , Humans , Macrophages/pathology , Magnetic Resonance Imaging , Mice , Rats , Rats, Transgenic
8.
Oncotarget ; 7(49): 80382-80390, 2016 Dec 06.
Article in English | MEDLINE | ID: mdl-27806344

ABSTRACT

Pleiotrophin (PTN) augments tumor growth by increasing proliferation of tumor cells and promoting vascular abnormalization, but its role in early gliomagenesis has not been evaluated. Through analysis of publically available datasets, we demonstrate that increased PTN mRNA expression is associated with amplification of chromosome 7, identified as one of the earliest steps in glioblastoma development. To elucidate the role of PTN in tumor initiation we employed the RCAS/tv-a model that allows glioma induction by RCAS-virus mediated expression of oncogenes in neural progenitor cells. Intracranial injection of RCAS-PTN did not induce glioma formation when administrated alone, but significantly enhanced RCAS-platelet derived growth factor (PDGF)B-induced gliomagenesis. PTN co-treatment augmented PDGFB-induced Akt activation in neural progenitor cells in vitro, and enhanced neural sphere size associated with increased proliferation. Our data indicates that PTN expression is associated with chromosome 7 gain, and that PTN enhances PDGFB-induced gliomagenesis by stimulating proliferation of neural progenitor cells.


Subject(s)
Brain Neoplasms/metabolism , Carrier Proteins/metabolism , Cell Proliferation/drug effects , Cytokines/metabolism , Glioblastoma/metabolism , Neoplastic Stem Cells/drug effects , Neural Stem Cells/drug effects , Proto-Oncogene Proteins c-sis/pharmacology , Animals , Avian Proteins/genetics , Avian Sarcoma Viruses/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Carrier Proteins/genetics , Cells, Cultured , Chromosomes, Human, Pair 7 , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cytokines/genetics , Gene Amplification , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mice, Transgenic , Neoplasm Grading , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Virus/genetics , Signal Transduction/drug effects , Spheroids, Cellular , Transfection
9.
Virus Genes ; 52(3): 365-71, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27108997

ABSTRACT

Transduction of oncogenes by ALVs and generation of acute transforming viruses is common in natural viral infections. In order to understand the molecular basis for the rapid oncogenicity of Fu-J, an acutely transforming avian leukosis virus isolated from fibrosarcomas in crossbreed broilers infected with subgroup J avian leukosis virus (ALV-J) in China, complete genomic structure of Fu-J virus was determined by PCR amplification and compared with those of Fu-J1, Fu-J2, Fu-J3, Fu-J4, and Fu-J5 reported previously. The results showed that the genome of Fu-J was defective, with parts of gag gene replaced by the complete v-fps oncogene and encoded a 137 kDa Gag-fps fusion protein. Sequence analysis revealed that Fu-J and Fu-J1 to Fu-J5 were related quasi-species variants carrying different lengths of v-fps oncogenes generated from recombination between helper virus and c-fps gene. Comparison of virus carrying v-fps oncogene also gave us a glimpse of the molecular characterization and evolution process of the acutely transforming ALV.


Subject(s)
Avian Leukosis Virus/genetics , Avian Leukosis/virology , Fusion Proteins, gag-onc/genetics , Oncogene Proteins/genetics , Oncogenic Viruses/genetics , Poultry Diseases/virology , Protein-Tyrosine Kinases/genetics , Animals , Avian Leukosis Virus/isolation & purification , Avian Leukosis Virus/pathogenicity , Avian Sarcoma Viruses/genetics , Base Sequence , Chick Embryo , Chickens/virology , DNA, Viral , Fibrosarcoma/virology , Gene Products, gag/genetics , Genes, Viral , Helper Viruses/genetics , Retroviridae/genetics , Virus Replication
10.
Cell Mol Life Sci ; 72(23): 4671-80, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26109426

ABSTRACT

Human induced pluripotent stem cells (hiPSC) differentiate into multiple cell types. Selective cell targeting is often needed for analyzing gene function by overexpressing proteins in a distinct population of hiPSC-derived cell types and for monitoring cell fate in response to stimuli. However, to date, this has not been possible, as commonly used viruses enter the hiPSC via ubiquitously expressed receptors. Here, we report for the first time the application of a heterologous avian receptor, the tumor virus receptor A (TVA), to selectively transduce TVA(+) cells in a mixed cell population. Expression of the TVA surface receptor via genetic engineering renders cells susceptible for infection by avian leucosis virus (ALV). We generated hiPSC lines with this stably integrated, ectopic TVA receptor gene that expressed the receptor while retaining pluripotency. The undifferentiated hiPSC(TVA+) as well as their differentiating progeny could be infected by recombinant ALV (so-called RCAS virus) with high efficiency. Due to incomplete receptor blocking, even sequential infection of differentiating or undifferentiated TVA(+) cells was possible. In conclusion, the TVA/RCAS system provides an efficient and gentle gene transfer system for hiPSC and extends our possibilities for selective cell targeting and lineage tracing studies.


Subject(s)
Avian Proteins/genetics , Avian Sarcoma Viruses/genetics , Genetic Engineering/methods , Induced Pluripotent Stem Cells/virology , Receptors, Virus/genetics , Avian Proteins/metabolism , Avian Sarcoma Viruses/pathogenicity , Cell Differentiation , Cell Lineage , Cells, Cultured , DNA Transposable Elements , Flow Cytometry/methods , Genetic Vectors , Humans , Induced Pluripotent Stem Cells/cytology , Receptors, Virus/metabolism
11.
Proc Natl Acad Sci U S A ; 112(22): E2947-56, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-25991858

ABSTRACT

Pathway-specific gene delivery is requisite for understanding complex neuronal systems in which neurons that project to different target regions are locally intermingled. However, conventional genetic tools cannot achieve simultaneous, independent gene delivery into multiple target cells with high efficiency and low cross-reactivity. In this study, we systematically screened all receptor-envelope pairs resulting from the combination of four avian sarcoma leukosis virus (ASLV) envelopes (EnvA, EnvB, EnvC, and EnvE) and five engineered avian-derived receptors (TVA950, TVB(S3), TVC, TVB(T), and DR-46TVB) in vitro. Four of the 20 pairs exhibited both high infection rates (TVA-EnvA, 99.6%; TVB(S3)-EnvB, 97.7%; TVC-EnvC, 98.2%; and DR-46TVB-EnvE, 98.8%) and low cross-reactivity (<2.5%). Next, we tested these four receptor-envelope pairs in vivo in a pathway-specific gene-transfer method. Neurons projecting into a limited somatosensory area were labeled with each receptor by retrograde gene transfer. Three of the four pairs exhibited selective transduction into thalamocortical neurons expressing the paired receptor (>98%), with no observed cross-reaction. Finally, by expressing three receptor types in a single animal, we achieved pathway-specific, differential fluorescent labeling of three thalamic neuronal populations, each projecting into different somatosensory areas. Thus, we identified three orthogonal pairs from the list of ASLV subgroups and established a new vector system that provides a simultaneous, independent, and highly specific genetic tool for transferring genes into multiple target cells in vivo. Our approach is broadly applicable to pathway-specific labeling and functional analysis of diverse neuronal systems.


Subject(s)
Avian Sarcoma Viruses/genetics , Gene Transfer Techniques , Genetic Engineering/methods , Neural Pathways/cytology , Receptors, Virus/metabolism , Viral Envelope Proteins , Animals , Flow Cytometry , HEK293 Cells , Humans , Microscopy, Fluorescence , Rats , Receptors, Virus/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
12.
Neuro Oncol ; 17(3): 361-71, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25416826

ABSTRACT

BACKGROUND: Malignant gliomas are complex systems containing a number of factors that drive tumor initiation and progression, including genetic aberrations that lead to extensive cellular heterogeneity within the neoplastic compartment. Mouse models recapitulate these genetic aberrations, but readily observable heterogeneity remains challenging. METHODS: To interrogate cellular heterogeneity in mouse glioma models, we utilized a replication-competent avian sarcoma-leukosis virus long terminal repeat with splice acceptor/tumor virus A (RCAS-tva) system to generate spontaneous mouse gliomas that contained a Sox2-enhanced green fluorescent protein (EGFP) reporter. Glial fibrillary acidic protein-tva mice were crossed with Sox2-EGFP mice, and tumors were initiated that contained a subpopulation of Sox2-EGFP-high cells enriched for tumor-initiating cell properties such as self-renewal, multilineage differentiation potential, and perivascular localization. RESULTS: Following implantation into recipient mice, Sox2-EGFP-high cells generated tumors containing Sox2-EGFP-high and Sox2-EGFP-low cells. Kinomic analysis of Sox2-EGFP-high cells revealed activation of known glioma signaling pathways that are strongly correlated with patient survival including platelet-derived growth factor receptor beta, phosphoinositide-3 kinase, and vascular endothelial growth factor. Our functional analysis identified active feline sarcoma (Fes) signaling in Sox2-EGFP-high cells. Fes negatively correlated with glioma patient survival and was coexpressed with Sox2-positive cells in glioma xenografts and primary patient-derived tissue. CONCLUSIONS: Our RCAS-tva/Sox2-EGFP model will empower closer examination of cellular heterogeneity and will be useful for identifying novel glioma pathways as well as testing preclinical treatment efficacy.


Subject(s)
Brain Neoplasms/pathology , Disease Models, Animal , Genes, Reporter , Glioma/pathology , Neoplastic Stem Cells/pathology , SOXB1 Transcription Factors/genetics , Animals , Avian Leukosis Virus/genetics , Avian Sarcoma Viruses/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/virology , Genetic Vectors , Glioma/genetics , Glioma/metabolism , Glioma/virology , Green Fluorescent Proteins/genetics , Humans , Mice , Mice, Transgenic , Neoplastic Stem Cells/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction , Tumor Cells, Cultured
13.
Cold Spring Harb Protoc ; 2014(11): 1167-9, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-25368308

ABSTRACT

Injection of RCAS viruses is highly customizable to the desired target tissue. RCAS viruses can be delivered into mice in vivo by injection of virus-producing cells or by injection of concentrated virus. When cells are injected, they persist for several days, continuously producing virus. Typically the decision of whether to inject virus-producing cells or concentrated virus is determined by the volume that can be reliably injected into a given tissue and the age of the animal when the virus delivery is performed. This general protocol describes the intraperitoneal injection of RCAS-expressing cells into mice and discusses the circumstances in which the injection of concentrated virus is preferred.


Subject(s)
Avian Sarcoma Viruses/growth & development , Avian Sarcoma Viruses/genetics , Cell Transplantation/methods , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Animals , Injections, Intraperitoneal , Mice
14.
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
15.
J Gen Virol ; 95(Pt 9): 2060-2070, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24903328

ABSTRACT

Ancient endogenous retroviruses (ERVs), designated endogenous avian retrovirus (EAVs), are present in all Gallus spp. including the chicken, and resemble the modern avian sarcoma and leukosis viruses (ASLVs). The EAVs comprise several distinct retroviruses, including EAV-0, EAV-E51 and EAV-HP, as well as a putative member previously named the avian retrotransposon of chickens (ART-CH). Thus far, only the EAV-HP elements have been well characterized. Here, we determined sequences of representative EAV-0 and EAV-E51 proviruses by cloning and data mining of the 2011 assembly of the Gallus gallus genome. Although the EAV-0 elements are primarily deleted in the env region, we identified two complete EAV-0 env genes within the G. gallus genome and prototype elements sharing identity with an EAV-E51-related clone previously designated EAV-E33. Prototype EAV-0, EAV-E51 and EAV-E33 gag, pol and env gene sequences used for phylogenetic analysis of deduced proteins showed that the EAVs formed three distinct clades, with EAV-0 sharing the last common ancestor with the ASLVs. The EAV-E51 clade showed the greatest level of divergence compared with other EAVs or ASLVs, suggesting that these ERVs represented exogenous retroviruses that evolved and integrated into the germline over a long period of time. Moreover, the degree of divergence between the chicken and red jungle fowl EAV-E51 sequences suggested that they were more ancient than the other EAVs and may have diverged through mutations that accumulated post-integration. Finally, we showed that the ART-CH elements were chimeric defective ERVs comprising portions of EAV-E51 and EAV-HP rather than authentic retrotransposons.


Subject(s)
Avian Leukosis Virus/genetics , Avian Sarcoma Viruses/genetics , Chickens/virology , Endogenous Retroviruses/genetics , Retroelements/genetics , Amino Acid Sequence , Animals , Base Sequence , Biological Evolution , Chickens/genetics , DNA, Viral/genetics , Databases, Nucleic Acid , Gene Products, gag/genetics , Genetic Variation , Genome, Viral/genetics , Molecular Sequence Data , Phylogeny , Sequence Alignment , Sequence Analysis, RNA
17.
J Virol ; 87(4): 2137-50, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23221555

ABSTRACT

Integrated retroviral DNA is subject to epigenetic transcriptional silencing at different frequencies. This process is mediated by repressive DNA methylation and histone modifications on viral chromatin. However, the detailed mechanisms by which retroviral silencing is initiated and maintained are not well understood. Using a model system in which avian sarcoma virus (ASV) DNA is epigenetically repressed in mammalian cells, we previously found that a cellular scaffolding protein, Daxx, acts as an antiretroviral factor that promotes epigenetic repression through recruitment of histone deacetylases (HDACs). Here we show that human Daxx protein levels are increased in response to retroviral infection and that Daxx acts at the time of infection to initiate epigenetic repression. Consistent with a rapid and active antiviral epigenetic response, we found that repressive histone marks and long terminal repeat (LTR) DNA methylation could be detected within 12 h to 3 days postinfection, respectively. Daxx was also found to be required for long-term ASV silencing maintenance and full viral DNA methylation, and it was physically associated with both viral DNA and DNA methyltransferases (DNMTs). These findings support a model in which incoming retroviral protein-DNA complexes are detected by Daxx, and the integrated provirus is rapidly chromatinized and repressed by DNA methylation and histone modification as part of an antiviral response. These results uncover a possible direct and active antiviral mechanism by which DNMTs can be recruited to retroviral DNA.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Avian Sarcoma Viruses/genetics , DNA Methylation , Epigenetic Repression , Gene Expression Regulation, Viral , Host-Pathogen Interactions , Nuclear Proteins/metabolism , Animals , Avian Sarcoma Viruses/physiology , Cell Line , Co-Repressor Proteins , Gene Silencing , Humans , Molecular Chaperones
18.
PLoS Pathog ; 8(5): e1002694, 2012.
Article in English | MEDLINE | ID: mdl-22589725

ABSTRACT

Disparate enveloped viruses initiate infection by fusing with endosomes. However, the highly diverse and dynamic nature of endosomes impairs mechanistic studies of fusion and identification of sub-cellular sites supporting the nucleocapsid release. We took advantage of the extreme stability of avian retrovirus-receptor complexes at neutral pH and of acid-dependence of virus-endosome fusion to isolate the latter step from preceding asynchronous internalization/trafficking steps. Viruses were trapped within endosomes in the presence of NH4Cl. Removal of NH4Cl resulted in a quick and uniform acidification of all subcellular compartments, thereby initiating synchronous viral fusion. Single virus imaging demonstrated that fusion was initiated within seconds after acidification and often culminated in the release of the viral core from an endosome. Comparative studies of cells expressing either the transmembrane or GPI-anchored receptor isoform revealed that the transmembrane receptor delivered the virus to more fusion-permissive compartments. Thus the identity of endosomal compartments, in addition to their acidity, appears to modulate viral fusion. A more striking manifestation of the virus delivery to distinct compartments in the presence of NH4Cl was the viral core release into the cytosol of cells expressing the transmembrane receptor and into endosomes of cells expressing the GPI-anchored isoform. In the latter cells, the newly released cores exhibited restricted mobility and were exposed to a more acidic environment than the cytoplasm. These cores appear to enter into the cytosol after an additional slow temperature-dependent step. We conclude that the NH4Cl block traps the virus within intralumenal vesicles of late endosomes in cells expressing the GPI-anchored receptor. Viruses surrounded by more than one endosomal membrane release their core into the cytoplasm in two steps--fusion with an intralumenal vesicle followed by a yet unknown temperature-dependent step that liberates the core from late endosomes.


Subject(s)
Avian Sarcoma Viruses/genetics , Avian Sarcoma Viruses/metabolism , Endosomes/virology , Viral Core Proteins/metabolism , Viral Fusion Proteins/metabolism , Ammonium Chloride/chemistry , Animals , Cell Compartmentation , Cell Line , Chlorocebus aethiops , Endosomes/metabolism , HEK293 Cells , Humans , Protein Isoforms/biosynthesis , Protein Transport , Retroviridae Proteins/genetics , Retroviridae Proteins/metabolism , Viral Core Proteins/genetics , Viral Fusion Proteins/genetics , Virus Internalization
19.
PLoS One ; 6(12): e27751, 2011.
Article in English | MEDLINE | ID: mdl-22145019

ABSTRACT

BACKGROUND: We applied crosslinking techniques as a first step in preparation of stable avian sarcoma virus (ASV) integrase (IN)-DNA complexes for crystallographic investigations. These results were then compared with the crystal structures of the prototype foamy virus (PFV) intasome and with published data for other retroviral IN proteins. METHODOLOGY/RESULTS: Photoaffinity crosslinking and site-directed chemical crosslinking were used to localize the sites of contacts with DNA substrates on the surface of ASV IN. Sulfhydryl groups of cysteines engineered into ASV IN and amino-modified nucleotides in DNA substrates were used for attachment of photocrosslinkers. Analysis of photocrosslinking data revealed several specific DNA-protein contacts. To confirm contact sites, thiol-modified nucleotides were introduced into oligo-DNA substrates at suggested points of contact and chemically crosslinked to the cysteines via formation of disulfide bridges. Cysteines incorporated in positions 124 and 146 in the ASV IN core domain were shown to interact directly with host and viral portions of the Y-mer DNA substrate, respectively. Crosslinking of an R244C ASV IN derivative identified contacts at positions 11 and 12 on both strands of viral DNA. The most efficient disulfide crosslinking was observed for complexes of the ASV IN E157C and D64C derivatives with linear viral DNA substrate carrying a thiol-modified scissile phosphate. CONCLUSION: Analysis of our crosslinking results as well as published results of retroviral IN protein from other laboratories shows good agreement with the structure of PFV IN and derived ASV, HIV, and MuLV models for the core domain, but only partial agreement for the N- and C-terminal domains. These differences might be explained by structural variations and evolutionary selection for residues at alternate positions to perform analogous functions, and by methodological differences: i.e., a static picture of a particular assembly from crystallography vs. a variety of interactions that might occur during formation of functional IN complexes in solution.


Subject(s)
Avian Sarcoma Viruses/enzymology , Cross-Linking Reagents/pharmacology , DNA, Viral/chemistry , DNA, Viral/metabolism , Integrases/chemistry , Integrases/metabolism , Amino Acid Sequence , Avian Sarcoma Viruses/genetics , Base Sequence , Binding Sites , Crystallography, X-Ray , DNA, Viral/genetics , Integrases/genetics , Light , Models, Chemical , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation/genetics , Protein Conformation , Sequence Homology, Amino Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
20.
J Biol Chem ; 286(29): 25710-8, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21622554

ABSTRACT

In the initial step of integration, retroviral integrase (IN) introduces precise nicks in the degenerate, short inverted repeats at the ends of linear viral DNA. The scissile phosphodiester bond is located immediately 3' of a highly conserved CA/GT dinucleotide, usually 2 bp from the ends. These nicks create new recessed 3'-OH viral DNA ends that are required for joining to host cell DNA. Previous studies have indicated that unpairing, "fraying," of the viral DNA ends by IN contributes to end recognition or catalysis. Here, we report that end fraying can be detected independently of catalysis with both avian sarcoma virus (ASV) and human immunodeficiency virus type 1 (HIV-1) IN proteins by use of fluorescence resonance energy transfer (FRET). The results were indicative of an IN-induced intramolecular conformational change in the viral DNA ends (cis FRET). Fraying activity is tightly coupled to the DNA binding capabilities of these enzymes, as follows: an inhibitor effective against both IN proteins was shown to block ASV IN DNA binding and end fraying, with similar dose responses; ASV IN substitutions that reduced DNA binding also reduced end fraying activity; and HIV-1 IN DNA binding and end fraying were both undetectable in the absence of a metal cofactor. Consistent with our previous results, end fraying is sequence-independent, suggesting that the DNA terminus per se is a major structural determinant for recognition. We conclude that frayed ends represent a functional intermediate in which DNA termini can be sampled for suitability for endonucleolytic processing.


Subject(s)
Avian Sarcoma Viruses/enzymology , Base Pairing , DNA, Viral/chemistry , HIV Integrase/metabolism , HIV-1/enzymology , Avian Sarcoma Viruses/genetics , Avian Sarcoma Viruses/metabolism , Base Sequence , Catalytic Domain , Coenzymes/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , Fluorescence Resonance Energy Transfer , HIV Integrase/chemistry , HIV-1/genetics , HIV-1/metabolism , Metals/metabolism , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...