Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 323
Filter
1.
Cancer Sci ; 112(12): 4920-4930, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34653294

ABSTRACT

BLNK (BASH/SLP-65) encodes an adaptor protein that plays an important role in B-cell receptor (BCR) signaling. Loss-of-function mutations in this gene are observed in human pre-B acute lymphoblastic leukemia (ALL), and a subset of Blnk knock-out (KO) mice develop pre-B-ALL. To understand the molecular mechanism of the Blnk mutation-associated pre-B-ALL development, retroviral tagging was applied to KO mice using the Moloney murine leukemia virus (MoMLV). The Blnk mutation that significantly accelerated the onset of MoMLV-induced leukemia and increased the incidence of pre-B-ALL Cebpb was identified as a frequent site of retroviral integration, suggesting that its upregulation cooperates with Blnk mutations. Transgenic expression of the liver-enriched activator protein (LAP) isoform of Cebpb reduced the number of mature B-lymphocytes in the bone marrow and inhibited differentiation at the pre-BI stage. Furthermore, LAP expression significantly accelerated leukemogenesis in Blnk KO mice and alone acted as a B-cell oncogene. Furthermore, an inverse relationship between BLNK and C/EBPß expression was also noted in human pre-B-ALL cases, and the high level of CEBPB expression was associated with short survival periods in patients with BLNK-downregulated pre-B-ALL. These results indicate the association between the C/EBPß transcriptional network and BCR signaling in pre-B-ALL development and leukemogenesis. This study gives insight into ALL progression and suggests that the BCR/C/EBPß pathway can be a therapeutic target.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , CCAAT-Enhancer-Binding Protein-beta/genetics , Moloney murine leukemia virus/physiology , Mutation , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Humans , Mice , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/virology , Up-Regulation , Virus Integration
2.
EMBO J ; 40(16): e106540, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34121210

ABSTRACT

Dendritic cells (DC) subsets, like Langerhans cells (LC), are immune cells involved in pathogen sensing. They express specific antimicrobial cellular factors that are able to restrict infection and limit further pathogen transmission. Here, we identify the alarmin S100A9 as a novel intracellular antiretroviral factor expressed in human monocyte-derived and skin-derived LC. The intracellular expression of S100A9 is decreased upon LC maturation and inversely correlates with enhanced susceptibility to HIV-1 infection of LC. Furthermore, silencing of S100A9 in primary human LC relieves HIV-1 restriction while ectopic expression of S100A9 in various cell lines promotes intrinsic resistance to both HIV-1 and MLV infection by acting on reverse transcription. Mechanistically, the intracellular expression of S100A9 alters viral capsid uncoating and reverse transcription. S100A9 also shows potent inhibitory effect against HIV-1 and MMLV reverse transcriptase (RTase) activity in vitro in a divalent cation-dependent manner. Our findings uncover an unexpected intracellular function of the human alarmin S100A9 in regulating antiretroviral immunity in Langerhans cells.


Subject(s)
Alarmins/genetics , Calgranulin B/genetics , HIV-1/physiology , Langerhans Cells/virology , Moloney murine leukemia virus/physiology , Retroviridae Infections/prevention & control , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cricetulus , HIV-1/genetics , Host-Pathogen Interactions , Humans , Langerhans Cells/immunology , Leukemia, Experimental/prevention & control , Mice , Moloney murine leukemia virus/genetics , Reverse Transcription , Transforming Growth Factor beta/immunology , Tumor Virus Infections/prevention & control , Virus Replication
3.
Blood ; 137(7): 908-922, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33174606

ABSTRACT

Epigenetic regulation is essential for the maintenance of the hematopoietic system, and its deregulation is implicated in hematopoietic disorders. In this study, UTX, a demethylase for lysine 27 on histone H3 (H3K27) and a component of COMPASS-like and SWI/SNF complexes, played an essential role in the hematopoietic system by globally regulating aging-associated genes. Utx-deficient (UtxΔ/Δ) mice exhibited myeloid skewing with dysplasia, extramedullary hematopoiesis, impaired hematopoietic reconstituting ability, and increased susceptibility to leukemia, which are the hallmarks of hematopoietic aging. RNA-sequencing (RNA-seq) analysis revealed that Utx deficiency converted the gene expression profiles of young hematopoietic stem-progenitor cells (HSPCs) to those of aged HSPCs. Utx expression in hematopoietic stem cells declined with age, and UtxΔ/Δ HSPCs exhibited increased expression of an aging-associated marker, accumulation of reactive oxygen species, and impaired repair of DNA double-strand breaks. Pathway and chromatin immunoprecipitation analyses coupled with RNA-seq data indicated that UTX contributed to hematopoietic homeostasis mainly by maintaining the expression of genes downregulated with aging via demethylase-dependent and -independent epigenetic programming. Of note, comparison of pathway changes in UtxΔ/Δ HSPCs, aged muscle stem cells, aged fibroblasts, and aged induced neurons showed substantial overlap, strongly suggesting common aging mechanisms among different tissue stem cells.


Subject(s)
Aging/genetics , Gene Expression Regulation/genetics , Hematopoiesis/genetics , Hematopoietic System/physiology , Histone Code/genetics , Histone Demethylases/physiology , Animals , Cellular Senescence/genetics , DNA Breaks, Double-Stranded , DNA Repair , Female , Genetic Predisposition to Disease , Hematopoiesis, Extramedullary , Histone Demethylases/deficiency , Histone Demethylases/genetics , Immune Reconstitution , Jumonji Domain-Containing Histone Demethylases/metabolism , Leukemia, Experimental/genetics , Leukemia, Experimental/virology , Male , Mice , Mice, Knockout , Moloney murine leukemia virus/physiology , Myeloid Cells/pathology , Radiation Chimera , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism , Transcription Factors/metabolism , Virus Integration
4.
Int J Mol Sci ; 20(21)2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31671637

ABSTRACT

Based on the up-regulation of the proviral integration site of the Moloney murine leukemia virus (Pim) kinase family (Pim1, 2, and 3) observed in several types of leukemias and lymphomas, the development of pan-Pim inhibitors is an attractive therapeutic strategy. While only PIM447 and AZD1208 have entered the clinical stages. To elucidate the interaction mechanisms of three Pim kinases with PIM447 and AZD1208, six Pim/ligand systems were studied by homology modeling, molecular docking, molecular dynamics (MD) simulation and molecular mechanics/generalized Born surface area (MM/GBSA) binding free energy calculation. The residues of the top group (Leu44, Val52, Ala65, Lys67, and Leu120 in Pim1) dominated the pan-Pim inhibitors binding to Pim kinases. The residues of the bottom group (Gln127, Asp128, and Leu174 in Pim1) were crucial for Pims/PIM447 systems, while the contributions of these residues were decreased sharply for Pims/AZD1208 systems. It is likely that the more potent pan-Pim inhibitors should be bound strongly to the top and bottom groups. The residues of the left, right and loop groups were located in the loop regions of the binding pocket, however, the flexibility of these regions triggered the protein interacting with diverse pan-Pim inhibitors efficiently. We hope this work can provide valuable information for the design of novel pan-Pim inhibitors in the future.


Subject(s)
Biphenyl Compounds/pharmacology , Moloney murine leukemia virus/physiology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/chemistry , Proto-Oncogene Proteins c-pim-1/metabolism , Thiazolidines/pharmacology , Binding Sites , Biphenyl Compounds/chemistry , Cell Line, Tumor , Cell Proliferation , Humans , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Protein Kinase Inhibitors/chemistry , Structural Homology, Protein , Structure-Activity Relationship , Thiazolidines/chemistry , Virus Attachment
5.
Viruses ; 11(7)2019 07 17.
Article in English | MEDLINE | ID: mdl-31319455

ABSTRACT

Moloney leukemia virus 10 (MOV10) is an RNA helicase that has been shown to affect the replication of several viruses. The effect of MOV10 on Hepatitis B virus (HBV) infection is not known and its role on the replication of this virus is poorly understood. We investigated the effect of MOV10 down-regulation and MOV10 over-expression on HBV in a variety of cell lines, as well as in an infection system using a replication competent virus. We report that MOV10 down-regulation, using siRNA, shRNA, and CRISPR/Cas9 gene editing technology, resulted in increased levels of HBV DNA, HBV pre-genomic RNA, and HBV core protein. In contrast, MOV10 over-expression reduced HBV DNA, HBV pre-genomic RNA, and HBV core protein. These effects were consistent in all tested cell lines, providing strong evidence for the involvement of MOV10 in the HBV life cycle. We demonstrated that MOV10 does not interact with HBV-core. However, MOV10 binds HBV pgRNA and this interaction does not affect HBV pgRNA decay rate. We conclude that the restriction of HBV by MOV10 is mediated through effects at the level of viral RNA.


Subject(s)
Hepatitis B virus/physiology , Hepatitis B/virology , Host-Pathogen Interactions , Microbial Interactions , Moloney murine leukemia virus/physiology , Virus Replication , Animals , Cell Line , Cells, Cultured , Gene Expression Regulation, Viral , Humans , Mice , Protein Binding , RNA , RNA Helicases/metabolism , RNA, Viral , Viral Proteins/metabolism
6.
Virology ; 529: 177-185, 2019 03.
Article in English | MEDLINE | ID: mdl-30716579

ABSTRACT

Variants of Ross River virus (RRV) that bind to heparan sulfate (HS) were previously selected by serial passaging in cell culture. To explore the effects of mutations that convey HS utilization, we pseudotyped Moloney murine leukemia virus (MoMLV), with the RRV envelope. We substituted amino-acid residues 216 and 218 on RRV-E2-envelope glycoprotein with basic amino-acid residues, because these mutations confer affinity for HS upon RRV. However, T216R-RRV- and N218R-RRV-pseudotyped viruses possessed lower transduction titers, and we demonstrated that HS-affinity impeded release of pseudotyped virus from producer cells. Addition of heparinase to HS-expressing target cells reduces the transduction efficiency of the T216R-RRV- and N218R-RRV-pseudotyped viruses, whereas no such effect is seen in cells lacking HS. Under appropriate conditions, these T216R-RRV- and N218R-RRV-pseudotyped viruses have enhanced capacities for transducing HS-expressing cells. General principles concerning viral adaptation to the use of attachment factors and design of pseudotyped viral vectors are discussed.


Subject(s)
Heparitin Sulfate/physiology , Moloney murine leukemia virus/physiology , Ross River virus/physiology , Viral Envelope Proteins/physiology , Virus Release/physiology , Animals , Cell Line , Cricetinae , Mice , Mutation , Protein Binding , Virus Internalization
7.
Biochem Biophys Res Commun ; 500(2): 365-369, 2018 06 02.
Article in English | MEDLINE | ID: mdl-29660337

ABSTRACT

Murine leukemia virus (MuLV) is a retrovirus known causing leukemia and neurological disorders in mice, and its viral life cycle and pathogenesis have been investigated extensively over the past decades. As a natural antiviral agent, betulinic acid is a pentacyclic triterpenoid that can be found in the bark of several species of plants (particularly the white birch). One of the hurdles for betulinic acid to release its antiviral potency is its poor water solubility. In this study, we synthesized more water-soluble ionic derivatives of betulinic acid, and examined their activities against Moloney MuLV (M-MuLV). The mouse fibroblast cells stably infected with M-MuLV, 43D cells, were treated with various doses of betulinic acids and its derivatives, and the viral structural protein Gag in cells and media were detected by western blots. Two ionic derivatives containing the benzalkonium cation were found to inhibit the virus production into media and decreased Gag in cells. However, a cell proliferation assay showed that the benzalkonium cation inhibited the growth of 43D cells, suggesting that our ionic derivatives limited virus production through the inhibition of metabolism in 43D cells. Interestingly, all of these betulinic acid compounds exhibited a minimum impact on the processing and release of Gag from 43D cells, which outlines the differences of viral maturation between MuLV and human immunodeficiency virus.


Subject(s)
Moloney murine leukemia virus/physiology , Triterpenes/pharmacology , Virus Replication/drug effects , Animals , Benzalkonium Compounds/pharmacology , Cell Death/drug effects , Cell Line , Ions , Mice , Moloney murine leukemia virus/drug effects , Pentacyclic Triterpenes , Triterpenes/chemistry , Betulinic Acid
8.
Virology ; 516: 165-175, 2018 03.
Article in English | MEDLINE | ID: mdl-29407374

ABSTRACT

Moloney Murine Leukemia Virus (M-MLV) proviral DNA is transcriptionally silenced in embryonic cells by a large repressor complex tethered to the provirus by two sequence-specific DNA binding proteins, ZFP809 and YY1. A central component of the complex is Trim28, a scaffold protein that regulates many target genes involved in cell cycle progression, DNA damage responses, and viral gene expression. The silencing activity of Trim28, and its interactions with corepressors are often regulated by post-translational modifications such as sumoylation and phosphorylation. We defined the interaction domains of Trim28 and YY1, and investigated the role of sumoylation and phosphorylation of Trim28 in mediating M-MLV silencing. The RBCC domain of Trim28 was sufficient for interaction with YY1, and acidic region 1 and zinc fingers of YY1 were necessary and sufficient for its interaction with Trim28. Additionally, we found that residue K779 was critical for Trim28-mediated silencing of M-MLV in embryonic cells.


Subject(s)
Moloney murine leukemia virus/physiology , Proviruses/genetics , Retroviridae Infections/veterinary , Rodent Diseases/metabolism , Tripartite Motif-Containing Protein 28/metabolism , YY1 Transcription Factor/metabolism , Amino Acid Motifs , Animals , Gene Silencing , Mice , Moloney murine leukemia virus/genetics , Protein Binding , Protein Domains , Proviruses/metabolism , Retroviridae Infections/genetics , Retroviridae Infections/metabolism , Retroviridae Infections/virology , Rodent Diseases/genetics , Rodent Diseases/virology , Tripartite Motif-Containing Protein 28/chemistry , Tripartite Motif-Containing Protein 28/genetics , YY1 Transcription Factor/chemistry , YY1 Transcription Factor/genetics
9.
J Virol ; 91(12)2017 06 15.
Article in English | MEDLINE | ID: mdl-28381565

ABSTRACT

The relative contributions of cell-free virion circulation and direct cell-to-cell transmission to retroviral dissemination and pathogenesis are unknown. Tetherin/Bst2 is an antiviral protein that blocks enveloped virion release into the extracellular milieu but may not inhibit cell-to-cell virus transmission. We developed live-cell imaging assays which show that tetherin does not affect Moloney murine leukemia virus (MoMLV) spread, and only minimally affects vesicular stomatitis virus (VSV) spread, to adjacent cells in a monolayer. Conversely, cell-free MLV and VSV virion yields and VSV spread to distal cells were dramatically reduced by tetherin. To elucidate the roles of tetherin and cell-free virions during in vivo viral dissemination and pathogenesis, we developed mice carrying an inducible human tetherin (hTetherin) transgene. While ubiquitous hTetherin expression was detrimental to the growth and survival of mice, restriction of hTetherin expression to hematopoietic cells gave apparently healthy mice. The expression of hTetherin in hematopoietic cells had little or no effect on the number of MoMLV-infected splenocytes and thymocytes. However, hTetherin expression significantly reduced cell-free plasma viremia and also delayed MoMLV-induced disease. Overall, these results suggest that MoMLV spread within hematopoietic tissues and cell monolayers involves cell-to-cell transmission that is resistant to tetherin but that virion dissemination via plasma is inhibited by tetherin and is required for full MoMLV pathogenesis.IMPORTANCE Retroviruses are thought to spread primarily via direct cell-to-cell transmission, yet many have evolved to counteract an antiviral protein called tetherin, which may selectively inhibit cell-free virus release. We generated a mouse model with an inducible tetherin transgene in order to study how tetherin affects retroviral dissemination and on which cell types its expression is required to do so. We first developed a novel in vitro live-cell imaging assay to demonstrate that while tetherin does indeed dramatically reduce cell-free virus spreading, it has little to no effect on direct cell-to-cell transmission of either vesicular stomatitis virus (VSV) or the retrovirus MoMLV. Using our transgenic mouse model, we found that tetherin expression on hematopoietic cells resulted in the specific reduction of MoMLV cell-free plasma viremia but not the number of infected hematopoietic cells. The delay in disease associated with this scenario suggests a role for cell-free virus in retroviral disease progression.


Subject(s)
Antigens, CD/metabolism , Moloney murine leukemia virus/physiology , Retroviridae Infections/virology , Vesicular stomatitis Indiana virus/physiology , Virus Internalization , Virus Release , Animals , Antigens, CD/genetics , Antigens, CD/pharmacology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/pharmacology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/virology , Humans , Mice , Mice, Transgenic , NIH 3T3 Cells , Spleen/cytology , Spleen/virology , Thymocytes/virology , Viremia , Virion/metabolism , Virus Replication
10.
Sci Rep ; 7: 41769, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28150748

ABSTRACT

Moloney murine leukemia virus reverse transcriptase (MMLV-RT) is a widely used enzyme for cDNA synthesis. Here we show that MMLV-RT has a strong template-independent polymerase activity using blunt DNA ends as substrate that generates 3' overhangs of A, C, G, or T. Nucleotides were appended efficiently in the order A > G > T > C, and tail lengths varied from 4 to 5, 2 to 7, 2 to 4, and 2 to 3 for A, C, G, and T, respectively. The activity was so strong that nearly all our test DNA ends were appended with at least one A, C, G, or T. The N-tailing activity of MMLV-RT was enhanced in the presence of Mn2+, and the G-, C-, and T-tailing activities were further enhanced by dCMP, dGMP, and dAMP, respectively. This is the first report of an enzymatic activity that almost thoroughly appends two or more As, or one or more Cs, Gs, or Ts to the 3' end of double-stranded DNA, which would enable exhaustive analysis of DNA samples. The N-tailing activity of MMLV-RT is potentially useful in many biotechnological applications.


Subject(s)
DNA/metabolism , Moloney murine leukemia virus/physiology , RNA-Directed DNA Polymerase/metabolism , Reverse Transcription , Animals , DNA/genetics , DNA, Complementary/genetics , DNA, Complementary/metabolism , Enzyme Activation , Mice , Moloney murine leukemia virus/genetics , Templates, Genetic
11.
Proc Natl Acad Sci U S A ; 114(6): E922-E930, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28115710

ABSTRACT

Replication of the murine leukemia viruses is strongly suppressed in mouse embryonic stem (ES) cells. Proviral DNAs are formed normally but are then silenced by a large complex bound to DNA by the ES cell-specific zinc-finger protein ZFP809. We show here that ZFP809 expression is not regulated by transcription but rather by protein turnover: ZFP809 protein is stable in embryonic cells but highly unstable in differentiated cells. The protein is heavily modified by the accumulation of polyubiquitin chains in differentiated cells and stabilized by the proteasome inhibitor MG132. A short sequence of amino acids at the C terminus of ZFP809, including a single lysine residue (K391), is required for the rapid turnover of the protein. The silencing cofactor TRIM28 was found to promote the degradation of ZFP809 in differentiated cells. These findings suggest that the stem cell state is established not only by an unusual transcriptional profile but also by unusual regulation of protein levels through the proteasomal degradation pathway.


Subject(s)
DNA-Binding Proteins/genetics , Gene Silencing , Moloney murine leukemia virus/genetics , Mouse Embryonic Stem Cells/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , DNA-Binding Proteins/metabolism , Gene Expression Regulation , HEK293 Cells , Host-Pathogen Interactions , Humans , Mice , Moloney murine leukemia virus/physiology , Mouse Embryonic Stem Cells/virology , NIH 3T3 Cells , Protein Binding , Proviruses/genetics , Proviruses/physiology , Retroviridae/genetics , Retroviridae/physiology , Tripartite Motif-Containing Protein 28/genetics , Tripartite Motif-Containing Protein 28/metabolism
12.
Acta Virol ; 60(4): 372-378, 2016.
Article in English | MEDLINE | ID: mdl-27928916

ABSTRACT

Tetherin (also referred to as BST-2 or CD317) is an antiviral cellular restriction factor that inhibits the release of many enveloped viruses. It is a 30-36 kDa type II transmembrane protein, expression of which is induced by type I interferon. Mouse tetherin inhibits nascent cell-free particle release. However, it is unclear whether mouse tetherin restricts cell-to-cell spread of moloney murine leukemia virus (Mo-MLV) or whether is the mouse tetherin involved in syncytium formation. To examine cell-to-cell spread and syncytium formation of Mo-MLV in the presence or absence of mouse tetherin, R peptide (the cytoplasmic tail of the transmembrane protein (TM); 16 amino acids) truncated Env expressing vector was constructed. It contained enhanced green fluorescent protein (EGFP) in the proline rich region (PRR) of Env. This R(-)Env full-length molecular clone could rule out virus-cell transmission due to the slightly reduced R(-)Env protein incorporation into the viral particles. When NIH3T3 cells stably expressing mouse tetherin were transfected with R(-)Env full-length molecular clone, syncytium formation was significantly enhanced in the tetherin-expressing cells. These data suggest that tetherin-mediated retention of R-defective virions on the cell surface could enhance syncytium formation. In addition, we found that the R(-)Env full-length molecular clone containing EGFP in the PRR of Env to be a useful tool allowing fast and convenient detection of syncytia by fluorescence microscopy.


Subject(s)
Antigens, CD/metabolism , Giant Cells/virology , Membrane Glycoproteins/metabolism , Moloney murine leukemia virus/physiology , Retroviridae Infections/veterinary , Rodent Diseases/metabolism , Animals , Antigens, CD/genetics , Giant Cells/metabolism , Membrane Glycoproteins/genetics , Mice , Moloney murine leukemia virus/genetics , NIH 3T3 Cells , Retroviridae Infections/genetics , Retroviridae Infections/metabolism , Retroviridae Infections/virology , Rodent Diseases/genetics , Rodent Diseases/virology , Virion/genetics , Virion/physiology , Virus Release
13.
J Virol ; 90(10): 5205-5209, 2016 May 15.
Article in English | MEDLINE | ID: mdl-26962222

ABSTRACT

Recent advances in fluorescence microscopy allow three-dimensional analysis of HIV-1 preintegration complexes in the nuclei of infected cells. To extend this investigation to gammaretroviruses, we engineered a fluorescent Moloney murine leukemia virus (MLV) system consisting of MLV-integrase fused to enhanced green fluorescent protein (MLV-IN-EGFP). A comparative analysis of lentiviral (HIV-1) and gammaretroviral (MLV) fluorescent complexes in the nuclei of infected cells revealed their different spatial distributions. This research tool has the potential to achieve new insight into the nuclear biology of these retroviruses.


Subject(s)
Cell Nucleus/ultrastructure , Cell Nucleus/virology , HIV-1/physiology , Moloney murine leukemia virus/physiology , Animals , Green Fluorescent Proteins/genetics , HIV-1/genetics , HIV-1/ultrastructure , HeLa Cells , Humans , Integrases/genetics , Mice , Microscopy, Fluorescence , Moloney murine leukemia virus/ultrastructure , Virus Integration
14.
Antimicrob Agents Chemother ; 60(4): 2195-208, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26810656

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) capsid plays crucial roles in HIV-1 replication and thus represents an excellent drug target. We developed a high-throughput screening method based on a time-resolved fluorescence resonance energy transfer (HTS-TR-FRET) assay, using the C-terminal domain (CTD) of HIV-1 capsid to identify inhibitors of capsid dimerization. This assay was used to screen a library of pharmacologically active compounds, composed of 1,280in vivo-active drugs, and identified ebselen [2-phenyl-1,2-benzisoselenazol-3(2H)-one], an organoselenium compound, as an inhibitor of HIV-1 capsid CTD dimerization. Nuclear magnetic resonance (NMR) spectroscopic analysis confirmed the direct interaction of ebselen with the HIV-1 capsid CTD and dimer dissociation when ebselen is in 2-fold molar excess. Electrospray ionization mass spectrometry revealed that ebselen covalently binds the HIV-1 capsid CTD, likely via a selenylsulfide linkage with Cys198 and Cys218. This compound presents anti-HIV activity in single and multiple rounds of infection in permissive cell lines as well as in primary peripheral blood mononuclear cells. Ebselen inhibits early viral postentry events of the HIV-1 life cycle by impairing the incoming capsid uncoating process. This compound also blocks infection of other retroviruses, such as Moloney murine leukemia virus and simian immunodeficiency virus, but displays no inhibitory activity against hepatitis C and influenza viruses. This study reports the use of TR-FRET screening to successfully identify a novel capsid inhibitor, ebselen, validating HIV-1 capsid as a promising target for drug development.


Subject(s)
Anti-HIV Agents/pharmacology , Azoles/pharmacology , Capsid Proteins/antagonists & inhibitors , Capsid/drug effects , HIV-1/drug effects , Organoselenium Compounds/pharmacology , Small Molecule Libraries/pharmacology , Anti-HIV Agents/chemistry , Azoles/chemistry , Binding Sites , Capsid/chemistry , Capsid/metabolism , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Databases, Pharmaceutical , Fluorescence Resonance Energy Transfer , HIV-1/physiology , HeLa Cells , High-Throughput Screening Assays , Humans , Isoindoles , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/virology , Moloney murine leukemia virus/drug effects , Moloney murine leukemia virus/physiology , Organoselenium Compounds/chemistry , Protein Binding , Protein Domains , Protein Multimerization/drug effects , Protein Structure, Secondary , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/physiology , Small Molecule Libraries/chemistry , Virus Assembly/drug effects , Virus Assembly/physiology , Virus Replication/drug effects
15.
J Biol Chem ; 290(45): 27345-27359, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26370074

ABSTRACT

Siglec-1 (sialoadhesin, CD169) is a surface receptor on human cells that mediates trans-enhancement of HIV-1 infection through recognition of sialic acid moieties in virus membrane gangliosides. Here, we demonstrate that mouse Siglec-1, expressed on the surface of primary macrophages in an interferon-α-responsive manner, captures murine leukemia virus (MLV) particles and mediates their transfer to proliferating lymphocytes. The MLV infection of primary B-cells was markedly more efficient than that of primary T-cells. The major structural protein of MLV particles, Gag, frequently co-localized with Siglec-1, and trans-infection, primarily of surface-bound MLV particles, efficiently occurred. To explore the role of sialic acid for MLV trans-infection at a submolecular level, we analyzed the potential of six sialic acid precursor analogs to modulate the sialylated ganglioside-dependent interaction of MLV particles with Siglec-1. Biosynthetically engineered sialic acids were detected in both the glycolipid and glycoprotein fractions of MLV producer cells. MLV released from cells carrying N-acyl-modified sialic acids displayed strikingly different capacities for Siglec-1-mediated capture and trans-infection; N-butanoyl, N-isobutanoyl, N-glycolyl, or N-pentanoyl side chain modifications resulted in up to 92 and 80% reduction of virus particle capture and trans-infection, respectively, whereas N-propanoyl or N-cyclopropylcarbamyl side chains had no effect. In agreement with these functional analyses, molecular modeling indicated reduced binding affinities for non-functional N-acyl modifications. Thus, Siglec-1 is a key receptor for macrophage/lymphocyte trans-infection of surface-bound virions, and the N-acyl side chain of sialic acid is a critical determinant for the Siglec-1/MLV interaction.


Subject(s)
Moloney murine leukemia virus/pathogenicity , Sialic Acid Binding Ig-like Lectin 1/chemistry , Sialic Acid Binding Ig-like Lectin 1/physiology , Animals , Binding Sites , Cell Line , Gangliosides/chemistry , Gangliosides/metabolism , Host-Pathogen Interactions/physiology , Humans , Interferon-alpha/physiology , Leukemia, Experimental/physiopathology , Leukemia, Experimental/virology , Lymphocytes/physiology , Lymphocytes/virology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/physiology , N-Acetylneuraminic Acid/chemistry , Receptors, Virus/chemistry , Receptors, Virus/physiology , Retroviridae Infections/physiopathology , Retroviridae Infections/virology , Sialic Acid Binding Ig-like Lectin 1/genetics , Tumor Virus Infections/physiopathology , Tumor Virus Infections/virology
16.
Methods Mol Biol ; 1290: 127-40, 2015.
Article in English | MEDLINE | ID: mdl-25740482

ABSTRACT

The ability to introduce DNA elements into host cells and analyze the effects has revolutionized modern biology. Here we describe a protocol to generate Moloney murine leukemia virus (MMLV)-based, replication-incompetent pseudotyped retrovirus capable of infecting axolotls and incorporating genetic information into their genome. When pseudotyped with vesicular stomatitis virus (VSV)-G glycoprotein, the retroviruses can infect a broad range of proliferative axolotl cell types. However, if the retrovirus is pseudotyped with an avian sarcoma leukosis virus (ASLV)-A envelope protein, only axolotl cells experimentally manipulated to express the cognate tumor virus A (TVA) receptor can be targeted by infections. These strategies enable robust transgene expression over many cell divisions, cell lineage tracing, and cell subtype targeting for gene expression.


Subject(s)
Ambystoma mexicanum/virology , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/physiology , Transfection/methods , Alpharetrovirus/genetics , Ambystoma mexicanum/embryology , Animals , Extremities/embryology , Genetic Vectors/genetics , HEK293 Cells , Humans , Open Reading Frames/genetics , Plasmids/genetics , Viral Load
17.
Biotechnol Lett ; 37(5): 991-1001, 2015 May.
Article in English | MEDLINE | ID: mdl-25650340

ABSTRACT

Ligation-mediated-PCR was performed followed by the mapping of 177 and 150 integration sites from HepG2 and Hek293 transduced with chimera vector carrying recombinant human Factor IX (rhFIX) cDNA, respectively. The sequences were analyzed for chromosome preference, CpG, transcription start site (TSS), repetitive elements, fragile sites and target genes. In HepG2, rhFIX was had an increased preference for chromosomes 6 and 17; the median distance to the nearest CpG islands was 15,240 base pairs and 37 % of the integrations occurred in RefSeq genes. In Hek293, rhFIX had an increased preference for chromosome 5; the median distance to the nearest CpG islands was 209,100 base pairs and 74 % of the integrations occurred in RefSeq genes. The integrations in both cell lines were distant from the TSS. The integration patterns associated with this vector are different in each cell line.


Subject(s)
Factor IX/genetics , Factor IX/metabolism , Moloney murine leukemia virus/physiology , Moloney murine sarcoma virus/physiology , Virus Integration , Cell Line , Genetic Vectors , Humans , Moloney murine leukemia virus/genetics , Moloney murine sarcoma virus/genetics , Polymerase Chain Reaction , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transduction, Genetic
18.
J Biol Chem ; 289(38): 26368-26382, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25107902

ABSTRACT

Integration, one of the hallmarks of retrovirus replication, is mediated by a nucleoprotein complex called the preintegration complex (PIC), in which viral DNA is associated with many protein components that are required for completion of the early phase of infection. A striking feature of the PIC is its powerful integration activity in vitro. The PICs from a freshly isolated cytoplasmic extract of infected cells are able to insert viral DNA into exogenously added target DNA in vitro. Therefore, a PIC-based in vitro assay is a reliable system for assessing protein factors influencing retroviral integration. In this study, we applied a microtiter plate-based in vitro assay to a screening study using a protein library that was produced by the wheat germ cell-free protein synthesis system. Using a library of human E3 ubiquitin ligases, we identified RFPL3 as a potential stimulator of human immunodeficiency virus, type 1 (HIV-1) PIC integration activity in vitro. This enhancement of PIC activity by RFPL3 was likely to be attributed to its N-terminal RING domain. To further understand the functional role of RFPL3 in HIV infection, we created a human cell line overexpressing RFPL3. Immunoprecipitation analysis revealed that RFPL3 was associated with the human immunodeficiency virus, type 1 PICs in infected cells. More importantly, single-round HIV-1 infection was enhanced significantly by RFPL3 expression. Our proteomic approach displays an advantage in the identification of new cellular proteins affecting the integration activity of the PIC and, therefore, contributes to the understanding of functional interaction between retroviral integration complexes and host factors.


Subject(s)
Carrier Proteins/physiology , HIV-1/physiology , Ubiquitin-Protein Ligases/physiology , HEK293 Cells , Humans , Moloney murine leukemia virus/physiology , Protein Binding , Titrimetry , Virus Integration
19.
Nucleic Acids Res ; 42(16): 10209-25, 2014.
Article in English | MEDLINE | ID: mdl-25147212

ABSTRACT

Retroviral replication proceeds through an obligate integrated DNA provirus, making retroviral vectors attractive vehicles for human gene-therapy. Though most of the host cell genome is available for integration, the process of integration site selection is not random. Retroviruses differ in their choice of chromatin-associated features and also prefer particular nucleotide sequences at the point of insertion. Lentiviruses including HIV-1 preferentially integrate within the bodies of active genes, whereas the prototypical gammaretrovirus Moloney murine leukemia virus (MoMLV) favors strong enhancers and active gene promoter regions. Integration is catalyzed by the viral integrase protein, and recent research has demonstrated that HIV-1 and MoMLV targeting preferences are in large part guided by integrase-interacting host factors (LEDGF/p75 for HIV-1 and BET proteins for MoMLV) that tether viral intasomes to chromatin. In each case, the selectivity of epigenetic marks on histones recognized by the protein tether helps to determine the integration distribution. In contrast, nucleotide preferences at integration sites seem to be governed by the ability for the integrase protein to locally bend the DNA duplex for pairwise insertion of the viral DNA ends. We discuss approaches to alter integration site selection that could potentially improve the safety of retroviral vectors in the clinic.


Subject(s)
HIV-1/physiology , Moloney murine leukemia virus/physiology , Virus Integration , Genetic Therapy , Genetic Vectors , HIV-1/enzymology , HIV-1/genetics , Humans , Integrases/chemistry , Integrases/metabolism , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Moloney murine leukemia virus/enzymology , Moloney murine leukemia virus/genetics
20.
J Virol ; 88(18): 10680-95, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24991018

ABSTRACT

UNLABELLED: Mammalian genomes are replete with retrotransposable elements, including endogenous retroviruses. DNA methyltransferase 3-like (DNMT3L) is an epigenetic regulator expressed in prospermatogonia, growing oocytes, and embryonic stem (ES) cells. Here, we demonstrate that DNMT3L enhances the interaction of repressive epigenetic modifiers, including histone deacetylase 1 (HDAC1), SET domain, bifurcated 1 (SETDB1), DNA methyltransferase 3A (DNMT3A), and tripartite motif-containing protein 28 (TRIM28; also known as TIF1ß and KAP1) in ES cells and orchestrates retroviral silencing activity with TRIM28 through mechanisms including, but not limited to, de novo DNA methylation. Ectopic expression of DNMT3L in somatic cells causes methylation-independent retroviral silencing activity by recruitment of the TRIM28/HDAC1/SETDB1/DNMT3A/DNMT3L complex to newly integrated Moloney murine leukemia virus (Mo-MuLV) proviral DNA. Concurrent with this recruitment, we also observed the accumulation of histone H3 lysine 9 trimethylation (H3K9me3) and heterochromatin protein 1 gamma (HP1γ), as well as reduced H3K9 and H3K27 acetylation at Mo-MuLV proviral sequences. Ectopic expression of DNMT3L in late-passage mouse embryonic fibroblasts (MEFs) recruited cytoplasmically localized HDAC1 to the nucleus. The formation of this epigenetic modifying complex requires interaction of DNMT3L with DNMT3A as well as with histone H3. In fetal testes at embryonic day 17.5, endogenous DNMT3L also enhanced the binding among TRIM28, DNMT3A, SETDB1, and HDAC1. We propose that DNMT3L may be involved in initiating a cascade of repressive epigenetic modifications by assisting in the preparation of a chromatin context that further attracts DNMT3A-DNMT3L binding and installs longer-term DNA methylation marks at newly integrated retroviruses. IMPORTANCE: Almost half of the mammalian genome is composed of endogenous retroviruses and other retrotransposable elements that threaten genomic integrity. These elements are usually subject to epigenetic silencing. We discovered that two epigenetic regulators that lack enzymatic activity, DNA methyltransferase 3-like (DNMT3L) and tripartite motif-containing protein 28 (TRIM28), collaborate with each other to impose retroviral silencing. In addition to modulating de novo DNA methylation, we found that by interacting with TRIM28, DNMT3L can attract various enzymes to form a DNMT3L-induced repressive complex to remove active marks and add repressive marks to histone proteins. Collectively, these results reveal a novel and pivotal function of DNMT3L in shaping the chromatin modifications necessary for retroviral and retrotransposon silencing.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Silencing , Leukemia, Experimental/enzymology , Leukemia, Experimental/genetics , Moloney murine leukemia virus/physiology , Repressor Proteins/metabolism , Animals , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Embryonic Stem Cells/enzymology , Embryonic Stem Cells/metabolism , Gene Expression , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histones/metabolism , Humans , Leukemia, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Moloney murine leukemia virus/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Repressor Proteins/genetics , Tripartite Motif-Containing Protein 28
SELECTION OF CITATIONS
SEARCH DETAIL
...