Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.696
Filter
Add more filters

Publication year range
1.
PLoS Pathog ; 20(1): e1011640, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38215165

ABSTRACT

Retroviral reverse transcription starts within the capsid and uncoating and reverse transcription are mutually dependent. There is still debate regarding the timing and cellular location of HIV's uncoating and reverse transcription and whether it occurs solely in the cytoplasm, nucleus or both. HIV can infect non-dividing cells because there is active transport of the preintegration complex (PIC) across the nuclear membrane, but Murine Leukemia Virus (MLV) is thought to depend on cell division for replication and whether MLV uncoating and reverse transcription is solely cytoplasmic has not been studied. Here, we used NIH3T3 and primary mouse dendritic cells to determine where the different stages of reverse transcription occur and whether cell division is needed for nuclear entry. Our data strongly suggest that in both NIH3T3 cells and dendritic cells (DCs), the initial step of reverse transcription occurs in the cytoplasm. However, we detected MLV RNA/DNA hybrid intermediates in the nucleus of dividing NIH3T3 cells and non-dividing DCs, suggesting that reverse transcription can continue after nuclear entry. We also confirmed that the MLV PIC requires cell division to enter the nucleus of NIH3T3 cells. In contrast, we show that MLV can infect non-dividing primary DCs, although integration of MLV DNA in DCs still required the viral p12 protein. Knockdown of several nuclear pore proteins dramatically reduced the appearance of integrated MLV DNA in DCs but not NIH3T3 cells. Additionally, MLV capsid associated with the nuclear pore proteins NUP358 and NUP62 during infection. These findings suggest that simple retroviruses, like the complex retrovirus HIV, gain nuclear entry by traversing the nuclear pore complex in non-mitotic cells.


Subject(s)
HIV Infections , Nuclear Pore Complex Proteins , Animals , Mice , Nuclear Pore Complex Proteins/genetics , NIH 3T3 Cells , Leukemia Virus, Murine/genetics , Viral Proteins , Capsid Proteins , Retroviridae , DNA , Dendritic Cells
2.
J Virol ; 97(5): e0193022, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37093008

ABSTRACT

Inbred mouse lines vary in their ability to mount protective antiretroviral immune responses, and even closely related strains can exhibit opposing phenotypes upon retroviral infection. Here, we found that 129S mice inherit a previously unknown mechanism for the production of anti-murine leukemia virus (MLV) antibodies and control of infection. The resistant phenotype in 129S1 mice is controlled by two dominant loci that are independent from known MLV resistance genes. We also show that production of anti-MLV antibodies in 129S7 mice, but not 129S1 mice, is independent of interferon gamma signaling. Thus, our data indicate that 129S mice inherit an unknown mechanism for control of MLV infection and demonstrate that there is genetic variability in 129S substrains that affects their ability to mount antiviral immune responses. IMPORTANCE Understanding the genetic basis for production of protective antiviral immune responses is crucial for the development of novel vaccines and adjuvants. Additionally, characterizing the genetic and phenotypic variability in inbred mice has implications for the selection of strains for targeted mutagenesis, choice of controls, and for broader understanding of the requirements for protective immunity.


Subject(s)
Mice, Inbred Strains , Retroviridae Infections , Animals , Mice , Immunity , Interferon-gamma , Leukemia Virus, Murine/genetics , Mice, Inbred Strains/genetics , Mice, Inbred Strains/immunology , Retroviridae Infections/immunology
3.
Retrovirology ; 20(1): 16, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37700325

ABSTRACT

BACKGROUND: The murine leukemia virus (MLV) has been a powerful model of pathogenesis for the discovery of genes involved in cancer. Its splice donor (SD')-associated retroelement (SDARE) is important for infectivity and tumorigenesis, but the mechanism remains poorly characterized. Here, we show for the first time that P50 protein, which is produced from SDARE, acts as an accessory protein that transregulates transcription and induces cell transformation. RESULTS: By infecting cells with MLV particles containing SDARE transcript alone (lacking genomic RNA), we show that SDARE can spread to neighbouring cells as shown by the presence of P50 in infected cells. Furthermore, a role for P50 in cell transformation was demonstrated by CCK8, TUNEL and anchorage-independent growth assays. We identified the integrase domain of P50 as being responsible for transregulation of the MLV promoter using luciferase assay and RTqPCR with P50 deleted mutants. Transcriptomic analysis furthermore revealed that the expression of hundreds of cellular RNAs involved in cancerogenesis were deregulated in the presence of P50, suggesting that P50 induces carcinogenic processes via its transcriptional regulatory function. CONCLUSION: We propose a novel SDARE-mediated mode of propagation of the P50 accessory protein in surrounding cells. Moreover, due to its transforming properties, P50 expression could lead to a cellular and tissue microenvironment that is conducive to cancer development.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Mice , Animals , Genomics , Leukemia Virus, Murine/genetics , Promoter Regions, Genetic , RNA
4.
J Virol ; 95(18): e0063421, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34190600

ABSTRACT

The host transmembrane protein SERINC5 is incorporated into viral particles and restricts infection by certain retroviruses. However, what motif of SERINC5 mediates this process remains elusive. By conducting mutagenesis analyses, we found that the substitution of phenylalanine with alanine at position 412 (F412A) resulted in a >75-fold reduction in SERINC5's restriction function. The F412A substitution also resulted in the loss of SERINC5's function to sensitize HIV-1 neutralization by antibodies recognizing the envelope's membrane proximal region. A series of biochemical analyses revealed that F412A showed steady-state protein expression, localization at the cellular membrane, and incorporation into secreted virus particles to a greater extent than in the wild type. Furthermore, introduction of several amino acid mutations at this position revealed that the aromatic side chains, including phenylalanine, tyrosine, and tryptophan, were required to maintain SERINC5 functions to impair the virus-cell fusion process and virion infectivity. Moreover, the wild-type SERINC5 restricted infection of lentiviruses pseudotyped with envelopes of murine leukemia viruses, simian immunodeficiency virus, and HIV-2, and F412A abrogated this function. Taken together, our results highlight the importance of the aromatic side chain at SERINC5 position 412 to maintain its restriction function against diverse retrovirus envelopes. IMPORTANCE The host protein SERINC5 is incorporated into progeny virions of certain retroviruses and restricts the infectivity of these viruses or sensitizes the envelope glycoprotein to a class of neutralizing antibodies. However, how and which part of SERINC5 engages with the diverse array of retroviral envelopes and exerts its antiretroviral functions remain elusive. During mutagenesis analyses, we eventually found that the single substitution of phenylalanine with alanine, but not with tyrosine or tryptophan, at position 412 (F412A) resulted in the loss of SERINC5's functions toward diverse retroviruses, whereas F412A showed steady-state protein expression, localization at the cellular membrane, and incorporation into progeny virions to a greater extent than the wild type. Results suggest that the aromatic side chain at position 412 of SERINC5 plays a critical role in mediating antiviral functions toward various retroviruses, thus providing additional important information regarding host and retrovirus interaction.


Subject(s)
Amino Acids, Aromatic/genetics , Cell Membrane/metabolism , HIV Infections/virology , HIV-1/pathogenicity , Leukemia Virus, Murine/pathogenicity , Membrane Proteins/metabolism , Mutation , HEK293 Cells , HIV Infections/genetics , HIV-1/genetics , Host-Pathogen Interactions , Humans , Leukemia Virus, Murine/genetics , Membrane Proteins/genetics , Virulence
5.
Immunology ; 164(1): 190-206, 2021 09.
Article in English | MEDLINE | ID: mdl-33987830

ABSTRACT

It is important to characterize novel proteins involved in T- and B-cell responses. Our previous study demonstrated that a novel protein, Mus musculus Gm40600, reduced the proliferation of Mus musculus plasmablast (PB)-like SP 2/0 cells and B-cell responses induced in vitro by LPS. In the present study, we revealed that Gm40600 directly promoted CD4+ T-cell responses to indirectly up-regulate B-cell responses. Importantly, we found that CD4+ T-cell responses, including T-cell activation and differentiation and cytokine production, were increased in Gm40600 transgenic (Tg) mice and were reduced in Gm40600 knockout (KO) mice. Finally, we demonstrated that Gm40600 promoted the Ahnak-mediated calcium signalling pathway by interacting with Ahnak to maintain a cytoplasmic lateral location of Ahnak in CD4+ T cells. Collectively, our data suggest that Gm40600 promotes CD4+ T-cell activation to up-regulate the B-cell response via interacting with Ahnak to promote the calcium signalling pathway. The results suggest that targeting Gm40600 may be a means to control CD4+ T-cell-related diseases.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Leukemia Virus, Murine/genetics , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Animals , Calcium Signaling , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Immunity, Humoral , Immunomodulation , Intracellular Signaling Peptides and Proteins/genetics , Lymphocyte Activation , Mice, Inbred C57BL , Mice, Knockout , RNA-Directed DNA Polymerase/genetics
6.
Eur J Immunol ; 50(10): 1591-1597, 2020 10.
Article in English | MEDLINE | ID: mdl-32470143

ABSTRACT

Mice bearing CT26 tumors can be cured by administration of L19-mIL12 or F8-mTNF, two antibody fusion proteins which selectively deliver their cytokine payload to the tumor. In both settings, cancer cures crucially depended on CD8+ T cells and the AH1 peptide (derived from the gp70 protein of the murine leukemia virus) acted as the main tumor-rejection antigen, with ∼50% of CD8+ T cells in the neoplastic mass being AH1-specific after therapy. In order to characterize the clonality of the T cell response, its phenotype, and activation status, we isolated CD8+ T cells from tumors and secondary lymphoid organs and submitted them to T cell receptor (TCR) and total mRNA sequencing. We found an extremely diverse repertoire of more than 40 000 unique TCR sequences, but the ten most abundant TCRs accounted for >60% of CD8+ T-cell clones in the tumor. AH1-specific TCRs were consistently found among the most abundant sequences. AH1-specific T cells in the tumor had a tissue-resident memory phenotype. Treatment with L19-mIL12 led to overexpression of IL-12 receptor and of markers of cell activation and proliferation. These data suggest that the antitumor response driven by antibody-cytokine fusions proceeds through an oligoclonal expansion and activation of tumor-infiltrating CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Colon/pathology , Colonic Neoplasms/therapy , Immunotherapy/methods , Leukemia Virus, Murine/genetics , Animals , Antigens, Neoplasm/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Disease Models, Animal , Female , Humans , Immunologic Memory , Interleukin-12/therapeutic use , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Peptides/genetics , Peptides/immunology , Receptors, Antigen, T-Cell/genetics , Retroviridae Proteins, Oncogenic/genetics , Retroviridae Proteins, Oncogenic/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
7.
J Virol ; 94(13)2020 06 16.
Article in English | MEDLINE | ID: mdl-32321818

ABSTRACT

RNA modifications play diverse roles in regulating RNA function, and viruses co-opt these pathways for their own benefit. While recent studies have highlighted the importance of N6-methyladenosine (m6A)-the most abundant mRNA modification-in regulating retrovirus replication, the identification and function of other RNA modifications in viral biology have been largely unexplored. Here, we characterized the RNA modifications present in a model retrovirus, murine leukemia virus (MLV), using mass spectrometry and sequencing. We found that 5-methylcytosine (m5C) is highly enriched in viral genomic RNA relative to uninfected cellular mRNAs, and we mapped at single-nucleotide resolution the m5C sites, which are located in multiple clusters throughout the MLV genome. Further, we showed that the m5C reader protein ALYREF plays an important role in regulating MLV replication. Together, our results provide a complete m5C profile in a virus and its function in a eukaryotic mRNA.IMPORTANCE Over 130 modifications have been identified in cellular RNAs, which play critical roles in many cellular processes, from modulating RNA stability to altering translation efficiency. One such modification, 5-methylcytosine, is relatively abundant in mammalian mRNAs, but its precise location and function are not well understood. In this study, we identified unexpectedly high levels of m5C in the murine leukemia virus RNA, precisely mapped its location, and showed that ALYREF, a reader protein that specifically recognizes m5C, regulates viral production. Together, our findings provide a high-resolution atlas of m5C in murine leukemia virus and reveal a functional role of m5C in viral replication.


Subject(s)
5-Methylcytosine/metabolism , Leukemia Virus, Murine/genetics , 5-Methylcytosine/physiology , Animals , DNA Methylation/genetics , Genome, Viral/genetics , HEK293 Cells , Humans , Leukemia Virus, Murine/metabolism , Leukemia Virus, Murine/pathogenicity , Methyltransferases/metabolism , Mice , NIH 3T3 Cells , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Stability , RNA, Messenger/genetics , RNA, Viral/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Retroviridae/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome/genetics , Virus Replication/genetics
8.
PLoS Pathog ; 15(12): e1008154, 2019 12.
Article in English | MEDLINE | ID: mdl-31815961

ABSTRACT

Murine leukemia virus (MLV) integrase (IN) lacking the C-terminal tail peptide (TP) loses its interaction with the host bromodomain and extraterminal (BET) proteins and displays decreased integration at promoter/enhancers and transcriptional start sites/CpG islands. MLV lacking the IN TP via an altered open reading frame was used to infect tumorigenesis mouse model (MYC/Runx2) animals to observe integration patterns and phenotypic effects, but viral passage resulted in the restoration of the IN TP through small deletions. Mice subsequently infected with an MLV IN lacking the TP coding sequence (TP-) showed an improved median survival by 15 days compared to wild type (WT) MLV infection. Recombination with polytropic endogenous retrovirus (ERV), Pmv20, was identified in seven mice displaying both fast and slow tumorigenesis, highlighting the strong selection within the mouse to maintain the full-length IN protein. Mapping the genomic locations of MLV in tumors from an infected mouse with no observed recombination with ERVs, TP-16, showed fewer integrations at TSS and CpG islands, compared to integrations observed in WT tumors. However, this mouse succumbed to the tumor in relatively rapid fashion (34 days). Analysis of the top copy number integrants in the TP-16 tumor revealed their proximity to known MLV common insertion site genes while maintaining the MLV IN TP- genotype. Furthermore, integration mapping in K562 cells revealed an insertion preference of MLV IN TP- within chromatin profile states associated with weakly transcribed heterochromatin with fewer integrations at histone marks associated with BET proteins (H3K4me1/2/3, and H3K27Ac). While MLV IN TP- showed a decreased overall rate of tumorigenesis compared to WT virus in the MYC/Runx2 model, MLV integration still occurred at regions associated with oncogenic driver genes independently from the influence of BET proteins, either stochastically or through trans-complementation by functional endogenous Gag-Pol protein.


Subject(s)
Carcinogenesis , Genetic Vectors/toxicity , Leukemia, Experimental , Retroviridae Infections , Tumor Virus Infections , Animals , Chromatin , Core Binding Factor Alpha 1 Subunit/genetics , Disease Models, Animal , Genes, myc , Humans , Integrases/metabolism , K562 Cells , Leukemia Virus, Murine/genetics , Mice , Mice, Transgenic , Virus Integration
9.
Proc Natl Acad Sci U S A ; 115(22): E4990-E4999, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29769329

ABSTRACT

The insulin-responsive 12-transmembrane transporter GLUT4 changes conformation between an inward-open state and an outward-open state to actively facilitate cellular glucose uptake. Because of the difficulties of generating conformational mAbs against complex and highly conserved membrane proteins, no reliable tools exist to measure GLUT4 at the cell surface, follow its trafficking, or detect the conformational state of the protein. Here we report the isolation and characterization of conformational mAbs that recognize the extracellular and intracellular domains of GLUT4, including mAbs that are specific for the inward-open and outward-open states of GLUT4. mAbs against GLUT4 were generated using virus-like particles to present this complex membrane protein in its native conformation and using a divergent host species (chicken) for immunization to overcome immune tolerance. As a result, the isolated mAbs recognize conformational epitopes on native GLUT4 in cells, with apparent affinities as high as 1 pM and with specificity for GLUT4 across the human membrane proteome. Epitope mapping using shotgun mutagenesis alanine scanning across the 509 amino acids of GLUT4 identified the binding epitopes for mAbs specific for the states of GLUT4 and allowed the comprehensive identification of the residues that functionally control the GLUT4 inward-open and outward-open states. The mAbs identified here will be valuable molecular tools for monitoring GLUT4 structure, function, and trafficking, for differentiating GLUT4 conformational states, and for the development of novel therapeutics for the treatment of diabetes.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Glucose Transporter Type 4/immunology , Glucose Transporter Type 4/metabolism , Vaccines, Virus-Like Particle/immunology , Animals , Chickens , Epitope Mapping , Glucose Transporter Type 4/chemistry , Glucose Transporter Type 4/genetics , HEK293 Cells , Humans , Leukemia Virus, Murine/genetics , Models, Molecular , Protein Domains , Vaccines, Virus-Like Particle/chemistry
10.
Proc Natl Acad Sci U S A ; 115(50): E11751-E11760, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30478053

ABSTRACT

Retroviruses assemble and bud from infected cells in an immature form and require proteolytic maturation for infectivity. The CA (capsid) domains of the Gag polyproteins assemble a protein lattice as a truncated sphere in the immature virion. Proteolytic cleavage of Gag induces dramatic structural rearrangements; a subset of cleaved CA subsequently assembles into the mature core, whose architecture varies among retroviruses. Murine leukemia virus (MLV) is the prototypical γ-retrovirus and serves as the basis of retroviral vectors, but the structure of the MLV CA layer is unknown. Here we have combined X-ray crystallography with cryoelectron tomography to determine the structures of immature and mature MLV CA layers within authentic viral particles. This reveals the structural changes associated with maturation, and, by comparison with HIV-1, uncovers conserved and variable features. In contrast to HIV-1, most MLV CA is used for assembly of the mature core, which adopts variable, multilayered morphologies and does not form a closed structure. Unlike in HIV-1, there is similarity between protein-protein interfaces in the immature MLV CA layer and those in the mature CA layer, and structural maturation of MLV could be achieved through domain rotations that largely maintain hexameric interactions. Nevertheless, the dramatic architectural change on maturation indicates that extensive disassembly and reassembly are required for mature core growth. The core morphology suggests that wrapping of the genome in CA sheets may be sufficient to protect the MLV ribonucleoprotein during cell entry.


Subject(s)
Capsid/chemistry , Capsid/ultrastructure , Leukemia Virus, Murine/chemistry , Leukemia Virus, Murine/ultrastructure , Amino Acid Sequence , Animals , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/ultrastructure , Cryoelectron Microscopy , Crystallography, X-Ray , Electron Microscope Tomography , Gene Products, gag/chemistry , Gene Products, gag/genetics , Gene Products, gag/ultrastructure , HEK293 Cells , HIV-1/chemistry , HIV-1/genetics , HIV-1/ultrastructure , Humans , Leukemia Virus, Murine/genetics , Mice , Models, Molecular , Protein Domains , Protein Structure, Quaternary , Sequence Homology, Amino Acid , Virion/chemistry , Virion/genetics , Virion/ultrastructure
11.
J Virol ; 93(11)2019 06 01.
Article in English | MEDLINE | ID: mdl-30894464

ABSTRACT

Viruses can incorporate foreign glycoproteins to form infectious particles through a process known as pseudotyping. However, not all glycoproteins are compatible with all viruses. Despite the fact that viral pseudotyping is widely used, what makes a virus/glycoprotein pair compatible is poorly understood. To study this, we chose to analyze a gammaretroviral glycoprotein (Env) whose compatibility with different viruses could be modulated through small changes in its cytoplasmic tail (CT). One form of this glycoprotein is compatible with murine leukemia virus (MLV) particles but incompatible with human immunodeficiency virus type 1 (HIV-1) particles, while the second is compatible with HIV-1 particles but not with MLV particles. To decipher the factors affecting virus-specific Env incompatibility, we characterized Env incorporation, maturation, cell-to-cell fusogenicity, and virus-to-cell fusogenicity of each Env. The HIV-1 particle incompatibility correlated with less efficient cleavage of the R peptide by HIV-1 protease. However, the MLV particle incompatibility was more nuanced. MLV incompatibility appeared to be caused by lack of incorporation into particles, yet incorporation could be restored by further truncating the CT or by using a chimeric MLV Gag protein containing the HIV-1 MA without fully restoring infectivity. The MLV particle incompatibility appeared to be caused in part by fusogenic repression in MLV particles through an unknown mechanism. This study demonstrates that the Env CT can dictate functionality of Env within particles in a virus-specific manner.IMPORTANCE Viruses utilize viral glycoproteins to efficiently enter target cells during infection. How viruses acquire viral glycoproteins has been studied to understand the pathogenesis of viruses and develop safer and more efficient viral vectors for gene therapies. The CTs of viral glycoproteins have been shown to regulate various stages of glycoprotein biogenesis, but a gap still remains in understanding the molecular mechanism of glycoprotein acquisition and functionality regarding the CT. Here, we studied the mechanism of how specific mutations in the CT of a gammaretroviral envelope glycoprotein distinctly affect infectivity of two different viruses. Different mutations caused failure of glycoproteins to function in a virus-specific manner due to distinct fusion defects, suggesting that there are virus-specific characteristics affecting glycoprotein functionality.


Subject(s)
Gammaretrovirus/genetics , Gene Products, env/genetics , Viral Envelope Proteins/genetics , Animals , Cell Line , Cell Membrane/metabolism , Gammaretrovirus/metabolism , Gene Products, env/metabolism , Gene Products, gag/genetics , HEK293 Cells , HIV-1/metabolism , Humans , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/metabolism , Mice , Mutation , Species Specificity , Virion/metabolism , Virus Assembly
12.
J Virol ; 94(1)2019 12 12.
Article in English | MEDLINE | ID: mdl-31619553

ABSTRACT

The matrix (MA) domain of HIV-1 Gag plays key roles in virus assembly by targeting the Gag precursor to the plasma membrane and directing the incorporation of the viral envelope (Env) glycoprotein into virions. The latter function appears to be in part dependent on trimerization of the MA domain of Gag during assembly, as disruption of the MA trimer interface impairs Env incorporation. Conversely, many MA mutations that impair Env incorporation can be rescued by compensatory mutations in the trimer interface. In this study, we sought to investigate further the biological significance of MA trimerization by isolating and characterizing compensatory mutations that rescue MA trimer interface mutants with severely impaired Env incorporation. By serially propagating MA trimerization-defective mutants in T cell lines, we identified a number of changes in MA, both within and distant from the trimer interface. The compensatory mutations located within or near the trimer interface restored Env incorporation and particle infectivity and permitted replication in culture. The structure of the MA lattice was interrogated by measuring the cleavage of the murine leukemia virus (MLV) transmembrane Env protein by the viral protease in MLV Env-pseudotyped HIV-1 particles bearing the MA mutations and by performing crystallographic studies of in vitro-assembled MA lattices. These results demonstrate that rescue is associated with structural alterations in MA organization and rescue of MA domain trimer formation. Our data highlight the significance of the trimer interface of the MA domain of Gag as a critical site of protein-protein interaction during HIV-1 assembly and establish the functional importance of trimeric MA for Env incorporation.IMPORTANCE The immature Gag lattice is a critical structural feature of assembling HIV-1 particles, which is primarily important for virion formation and release. While Gag forms a hexameric lattice, driven primarily by the capsid domain, the MA domain additionally trimerizes where three Gag hexamers meet. MA mutants that are defective for trimerization are deficient for Env incorporation and replication, suggesting a requirement for trimerization of the MA domain of Gag in Env incorporation. This study used a gain-of-function, forced viral evolution approach to rescue HIV-1 mutants that are defective for MA trimerization. Compensatory mutations that rescue virus replication do so by restoring Env incorporation and MA trimer formation. This study supports the importance of MA domain trimerization in HIV-1 replication and the potential of the trimer interface as a therapeutic target.


Subject(s)
HIV-1/genetics , Viral Matrix Proteins/chemistry , Virion/genetics , Virus Assembly , env Gene Products, Human Immunodeficiency Virus/chemistry , gag Gene Products, Human Immunodeficiency Virus/chemistry , Amino Acid Motifs , Amino Acid Substitution , Animals , Cell Line , Gene Expression , HIV-1/metabolism , HeLa Cells , Humans , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/metabolism , Mice , Models, Molecular , Mutation , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Protein Multimerization , T-Lymphocytes/virology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Virion/metabolism , Virus Replication , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/metabolism , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism
13.
Arch Virol ; 165(5): 1089-1097, 2020 May.
Article in English | MEDLINE | ID: mdl-32146506

ABSTRACT

Gibbon ape leukemia virus (GALV) can infect a wide variety of cells but fails to infect most cells derived from laboratory mice. Transduction of human hematopoietic stem cells with GALV retroviral vectors is more efficient than with amphotropic vectors. In this study, a Moloney murine leukemia virus-gibbon ape leukemia virus (MoMLV-GALV) vector was constructed by replacing the natural env gene of the full-length Moloney MLV genome with the GALV env gene. To monitor viral transmission by green fluorescent protein (GFP) expression, internal ribosomal entry site-enhanced GFP (IRES-EGFP) was positioned between the GALV env gene and the 3' untranslated region (3' UTR) to obtain pMoMLV-GALV-EGFP. The MoMLV-GALV-EGFP vector was able to replicate with high titer in TE671 human rhabdomyosarcoma cells and U-87 human glioma cells. To evaluate the potential of the MoMLV-GALV vector as a therapeutic agent, the gene for the fusogenic envelope G glycoprotein of vesicular stomatitis virus (VSV-G) was incorporated into the vector. Infection with the resulting MoMLV-GALV-VSV-G vector resulted in lysis of the U-87 cells due to syncytium formation. Syncytium formation was also observed in the transfected human prostate cancer cell line LNCaP after extended cultivation of cells. In addition, we deleted the GALV env gene from the MoMLV-GALV-VSV-G vector to improve viral genome stability. This MoMLV-VSV-G vector is also replication competent and induces syncytium formation in 293T, HT1080, TE671 and U-87 cells. These results suggest that replication of the MoMLV-GALV-VSV-G vector or MoMLV-VSV-G vector may directly lead to cytotoxicity. Therefore, the vectors developed in this study are potentially useful tools for cancer gene therapy.


Subject(s)
Genetic Vectors , Leukemia Virus, Gibbon Ape/growth & development , Leukemia Virus, Murine/growth & development , Vesiculovirus/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Replication , Animals , Cell Line , Genetic Therapy/methods , Humans , Leukemia Virus, Gibbon Ape/genetics , Leukemia Virus, Murine/genetics , Mice , Neoplasms/therapy , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombination, Genetic
14.
Biochem Biophys Res Commun ; 514(2): 538-544, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31060775

ABSTRACT

Successful HIV-1 infection and subsequent replication deeply depend on how the virus usurps the host cell machinery. Identification and functional characterization of these host factors may represent a critical strategy for developing novel anti-HIV-1 therapy. Here, expression cloning with a cDNA expression library identified as an inhibitor of HIV-1 infection, a carboxy-terminally truncated form of human POZ/BTB and AT-hook- containing Zinc finger protein 1 (PATZ1), a transcriptional regulatory factor implicated in development and cancer. Knockdown or knockout of endogenous PATZ1 revealed a supportive role of PATZ1 in HIV-1 infection, but not in transduction with murine leukemia virus-based retroviral vector. More specifically, knockdown or knockout of PATZ1 impaired the viral cDNA synthesis but not the entry process and expression of two PATZ1 isoforms in PATZ1-KO cells restored susceptibility to HIV-1 infection. These results indicate that PATZ1 plays an important role in HIV-1 infection.


Subject(s)
HIV-1/genetics , Host-Pathogen Interactions/genetics , Kruppel-Like Transcription Factors/genetics , Lymphocytes/virology , RNA, Viral/genetics , Repressor Proteins/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation , Gene Library , HEK293 Cells , HIV-1/metabolism , HIV-1/pathogenicity , Humans , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/metabolism , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/metabolism , Lymphocytes/pathology , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Viral/biosynthesis , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Signal Transduction , Vesiculovirus/genetics , Vesiculovirus/metabolism
15.
Development ; 143(10): 1788-99, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27013243

ABSTRACT

The H3K9me3-specific histone methyltransferase Setdb1 impacts on transcriptional regulation by repressing both developmental genes and retrotransposons. How impaired retrotransposon silencing may lead to developmental phenotypes is currently unclear. Here, we show that loss of Setdb1 in pro-B cells completely abrogates B cell development. In pro-B cells, Setdb1 is dispensable for silencing of lineage-inappropriate developmental genes. Instead, we detect strong derepression of endogenous murine leukemia virus (MLV) copies. This activation coincides with an unusual change in chromatin structure, with only partial loss of H3K9me3 and unchanged DNA methylation, but strongly increased H3K4me3. Production of MLV proteins leads to activation of the unfolded protein response pathway and apoptosis. Thus, our data demonstrate that B cell development depends on the proper repression of retrotransposon sequences through Setdb1.


Subject(s)
Apoptosis/genetics , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/metabolism , Retroelements/genetics , Unfolded Protein Response/genetics , Animals , Gene Expression Profiling , Gene Silencing , HEK293 Cells , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Leukemia Virus, Murine/genetics , Lysine/metabolism , Methylation , Mice , Repetitive Sequences, Nucleic Acid/genetics , Transcription, Genetic
16.
J Virol ; 92(6)2018 03 15.
Article in English | MEDLINE | ID: mdl-29298890

ABSTRACT

The glycosylated Gag protein (gPr80) of murine leukemia viruses (MLVs) has been shown to exhibit multiple roles in facilitating retrovirus release, infection, and resistance to host-encoded retroviral restriction factors, such as APOBEC3, SERINC3, and SERINC5. One way in which gPr80 helps MLVs to escape host innate immune restriction is by increasing capsid stability, a feature that protects viral replication intermediates from being detected by cytosolic DNA sensors. gPr80 also increases the resistance of MLVs to deamination and restriction by mouse APOBEC3 (mA3). How the gPr80 accessory protein, with its three N-linked glycosylation sites, contributes to these resistance mechanisms is still not fully understood. Here we further characterized the function of gPr80 and, more specifically, revealed that the asparagines targeted for glycosylation in gPr80 also contribute to capsid stability through their parallel involvement in the Pr65 Gag structural polyprotein. In fact, we demonstrate that sensitivity to deamination by the mA3 and human A3 proteins is directly linked to capsid stability. We also show that full-length gPr80 is detected in purified viruses. However, our results suggest that gPr80 is inserted in the NexoCcyto orientation of a type I integral membrane protein. Additionally, our experiments have revealed the existence of a large population of Env-deficient virus-like particles (VLPs) harboring gPr80 inserted in the opposite (NcytoCexo) polarity, which is typical of type II integral membrane proteins. Overall this study provides new insight into the complex nature of the MLV gPr80 accessory protein.IMPORTANCE Viruses have evolved numerous strategies to infect, spread in, and persist in their hosts. Here we analyze the details of how the MLV-encoded glycosylated Gag (gPr80) protein protects the virus from being restricted by host innate immune defenses. gPr80 is a variant of the structural Pr65 Gag protein with an 88-amino-acid extended leader sequence that directs the protein for translation and glycosylation in the endoplasmic reticulum. This study dissects the specific contributions of gPr80 glycans and capsid stability in helping the virus to infect cells, spread, and counteract the effects of the host intrinsic restriction factor APOBEC3. Overall this study provides further insight into the elusive role of the gPr80 protein.


Subject(s)
Cytidine Deaminase/metabolism , Gene Products, gag/metabolism , Leukemia Virus, Murine/metabolism , APOBEC Deaminases , Animals , Cell Line , Cytidine Deaminase/genetics , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , Gene Products, gag/genetics , Humans , Leukemia Virus, Murine/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , NIH 3T3 Cells
17.
J Virol ; 92(11)2018 06 01.
Article in English | MEDLINE | ID: mdl-29593034

ABSTRACT

The apolipoprotein B editing complex 3 (APOBEC3) proteins are potent retroviral restriction factors that are under strong positive selection, both in terms of gene copy number and sequence diversity. A common feature of all the members of the APOBEC3 family is the presence of one or two cytidine deamination domains, essential for cytidine deamination of retroviral reverse transcripts as well as packaging into virions. Several studies have indicated that human and mouse APOBEC3 proteins restrict retrovirus infection via cytidine deaminase (CD)-dependent and -independent means. To understand the relative contribution of CD-independent restriction in vivo, we created strains of transgenic mice on an APOBEC3 knockout background that express a deaminase-dead mouse APOBEC3 due to point mutations in both CD domains (E73Q/E253Q). Here, we show that the CD-dead APOBEC3 can restrict murine retroviruses in vivo Moreover, unlike the wild-type protein, the mutant APOBEC3 is not packaged into virions but acts only as a cell-intrinsic restriction factor that blocks reverse transcription by incoming viruses. Finally, we show that wild-type and CD-dead mouse APOBEC3 can bind to murine leukemia virus (MLV) reverse transcriptase. Our findings suggest that the mouse APOBEC3 cytidine deaminase activity is not required for retrovirus restriction.IMPORTANCE APOBEC3 proteins are important host cellular restriction factors essential for restricting retrovirus infection by causing mutations in the virus genome and by blocking reverse transcription. While both methods of restriction function in vitro, little is known about their role during in vivo infection. By developing transgenic mice with mutations in the cytidine deamination domains needed for enzymatic activity and interaction with viral RNA, we show that APOBEC3 proteins can still restrict in vivo infection by interacting with reverse transcriptase and blocking its activity. These studies demonstrate that APOBEC3 proteins have evolved multiple means for blocking retrovirus infection and that all of these means function in vivo.


Subject(s)
Cytidine Deaminase/genetics , Leukemia Virus, Murine/genetics , Retroviridae Infections/prevention & control , Reverse Transcriptase Inhibitors/metabolism , Reverse Transcription/genetics , Animals , Cell Line , Cytidine Deaminase/metabolism , Deamination/genetics , HEK293 Cells , Humans , Leukemia Virus, Murine/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Viral/genetics , RNA-Directed DNA Polymerase/genetics
18.
Biologicals ; 59: 29-36, 2019 May.
Article in English | MEDLINE | ID: mdl-30992161

ABSTRACT

The utilization of the current combination of in vitro, in vivo and PCR assays for the identification of adventitious viruses in production cells has a limited range of detection. While Next Generation Sequencing (NGS) has a broader breadth of detection, it is unable to differentiate sequences from replicating viruses versus background inert sequences. In order to improve NGS specificity, we have designed a new NGS approach which targets subsets of viral RNAs only synthesized during cell infection. In order to evaluate the performance of this approach for detecting low levels of adventitious viruses, we selected two difficult virus/cell systems. This included B95-8 cells persistently infected by Human herpesvirus 4 (HHV-4) and serially diluted into HHV-4 negative Ramos cells and Madin-Darby bovine kidney cells with an early infection produced via a low dose of Bovine viral diarrhea virus. We demonstrated that the sensitivity of our RNA NGS approach was equivalent to targeted PCR with an increased specificity for the detection of viral infection. We were also able to identify a previously undetected Murine Leukemia Virus contaminant in Ramos cells. Based on these results, we conclude that this new RNA NGS approach is suitable for conducting viral safety evaluations of cells.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , RNA, Viral/genetics , Sequence Analysis, RNA/methods , Viruses/genetics , Animals , Cattle , Cell Line , Cell Line, Tumor , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Humans , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/physiology , Mice , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Virus Diseases/diagnosis , Virus Diseases/virology , Viruses/classification
19.
Nucleic Acids Res ; 45(22): 12954-12962, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29165701

ABSTRACT

Retroviral reverse transcriptase catalyses the synthesis of an integration-competent dsDNA molecule, using as a substrate the viral RNA. Using optical tweezers, we follow the Murine Leukemia Virus reverse transcriptase as it performs strand-displacement polymerization on a template under mechanical force. Our results indicate that reverse transcriptase functions as a Brownian ratchet, with dNTP binding as the rectifying reaction of the ratchet. We also found that reverse transcriptase is a relatively passive enzyme, able to polymerize on structured templates by exploiting their thermal breathing. Finally, our results indicate that the enzyme enters the recently characterized backtracking state from the pre-translocation complex.


Subject(s)
Algorithms , DNA, Viral/chemistry , Leukemia Virus, Murine/enzymology , Models, Chemical , RNA, Viral/chemistry , RNA-Directed DNA Polymerase/chemistry , DNA, Viral/genetics , DNA, Viral/metabolism , Deoxyribonucleotides/genetics , Deoxyribonucleotides/metabolism , Kinetics , Leukemia Virus, Murine/genetics , Optical Tweezers , Polymerization , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Directed DNA Polymerase/genetics , RNA-Directed DNA Polymerase/metabolism , Templates, Genetic , Thermodynamics
20.
Proc Natl Acad Sci U S A ; 113(8): 2086-91, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26858406

ABSTRACT

The bromodomain and extraterminal domain (BET) protein family are promising therapeutic targets for a range of diseases linked to transcriptional activation, cancer, viral latency, and viral integration. Tandem bromodomains selectively tether BET proteins to chromatin by engaging cognate acetylated histone marks, and the extraterminal (ET) domain is the focal point for recruiting a range of cellular and viral proteins. BET proteins guide γ-retroviral integration to transcription start sites and enhancers through bimodal interaction with chromatin and the γ-retroviral integrase (IN). We report the NMR-derived solution structure of the Brd4 ET domain bound to a conserved peptide sequence from the C terminus of murine leukemia virus (MLV) IN. The complex reveals a protein-protein interaction governed by the binding-coupled folding of disordered regions in both interacting partners to form a well-structured intermolecular three-stranded ß sheet. In addition, we show that a peptide comprising the ET binding motif (EBM) of MLV IN can disrupt the cognate interaction of Brd4 with NSD3, and that substitutions of Brd4 ET residues essential for binding MLV IN also impair interaction of Brd4 with a number of cellular partners involved in transcriptional regulation and chromatin remodeling. This suggests that γ-retroviruses have evolved the EBM to mimic a cognate interaction motif to achieve effective integration in host chromatin. Collectively, our findings identify key structural features of the ET domain of Brd4 that allow for interactions with both cellular and viral proteins.


Subject(s)
Integrases/chemistry , Leukemia Virus, Murine/enzymology , Nuclear Proteins/chemistry , Protein Folding , Transcription Factors/chemistry , Viral Proteins/chemistry , Amino Acid Motifs , Cell Cycle Proteins , Humans , Integrases/genetics , Integrases/metabolism , Leukemia Virus, Murine/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship , Transcription Factors/genetics , Transcription Factors/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL