Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 320
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Genet ; 11(10): e1005571, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26451601

ABSTRACT

A novel form of copy number control (CNC) helps maintain a low number of Ty1 retrovirus-like transposons in the Saccharomyces genome. Ty1 produces an alternative transcript that encodes p22, a trans-dominant negative inhibitor of Ty1 retrotransposition whose sequence is identical to the C-terminal half of Gag. The level of p22 increases with copy number and inhibits normal Ty1 virus-like particle (VLP) assembly and maturation through interactions with full length Gag. A forward genetic screen for CNC-resistant (CNCR) mutations in Ty1 identified missense mutations in GAG that restore retrotransposition in the presence of p22. Some of these mutations map within a predicted UBN2 domain found throughout the Ty1/copia family of long terminal repeat retrotransposons, and others cluster within a central region of Gag that is referred to as the CNCR domain. We generated multiple alignments of yeast Ty1-like Gag proteins and found that some Gag proteins, including those of the related Ty2 elements, contain non-Ty1 residues at multiple CNCR sites. Interestingly, the Ty2-917 element is resistant to p22 and does not undergo a Ty1-like form of CNC. Substitutions conferring CNCR map within predicted helices in Ty1 Gag that overlap with conserved sequence in Ty1/copia, suggesting that p22 disturbs a central function of the capsid during VLP assembly. When hydrophobic residues within predicted helices in Gag are mutated, Gag level remains unaffected in most cases yet VLP assembly and maturation is abnormal. Gag CNCR mutations do not alter binding to p22 as determined by co-immunoprecipitation analyses, but instead, exclude p22 from Ty1 VLPs. These findings suggest that the CNCR alleles enhance retrotransposition in the presence of p22 by allowing productive Gag-Gag interactions during VLP assembly. Our work also expands the strategies used by retroviruses for developing resistance to Gag-like restriction factors to now include retrotransposons.


Subject(s)
Gene Dosage/genetics , Gene Products, gag/genetics , Retroelements/genetics , Alleles , Gene Products, gag/biosynthesis , Genome, Fungal , Saccharomyces cerevisiae/genetics
2.
Med Sci Monit ; 23: 505-512, 2017 Jan 28.
Article in English | MEDLINE | ID: mdl-28130554

ABSTRACT

BACKGROUND Morphea, also known as localized scleroderma, is a rare autoimmune connective tissue disease characterized by skin fibrosis. UVA1 phototherapy is an important asset in the reduction of clinical manifestations in morphea. There are studies claiming that UV light modulates the expression of some human endogenous retroviral sequences. The aim of this study was to determine if the expression of HERV-K10 gag element is lowered by UVA1 phototherapy in morphea, a disease in which such irradiation has a soothing effect. MATERIAL AND METHODS The expression levels of the HERV-K10 gag were assessed by real-time PCR (polymerase chain reaction) in peripheral blood mononuclear cells (PBMC) and skin-punch biopsies of healthy volunteers and 9 morphea patients before and after phototherapy. Additionally, correlations between the HERV-K10 gag expression and age, disease duration, the Localized Scleroderma Skin Severity Index (LoSSI), and antinuclear antibody (ANA) titers were assessed. RESULTS In PBMC, HERV-K10 gag mRNA was significantly elevated after UVA1 phototherapy compared to healthy controls. Most of the patients responded with an increased expression level of this sequence. However, we found no statistical evidence at this point that phototherapy indeed has an effect on the HERV-K10 gag expression (there were no statistical differences in PBMC of morphea patients before and after phototherapy). Similarly, there was no statistically relevant effect of the UVA1 on the expression of HERV-K10 gag in skin. CONCLUSIONS At this point, the effect of UVA1 phototherapy on the expression of HERV-K10 gag cannot be statistically confirmed.


Subject(s)
Endogenous Retroviruses/radiation effects , Gene Products, gag/biosynthesis , Retroviridae Infections/therapy , Scleroderma, Localized/therapy , Ultraviolet Therapy/methods , Adult , Aged , Case-Control Studies , Endogenous Retroviruses/genetics , Endogenous Retroviruses/metabolism , Female , Gene Products, gag/genetics , Gene Products, gag/metabolism , Humans , Leukocytes, Mononuclear/radiation effects , Male , Middle Aged , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retroviridae Infections/blood , Retroviridae Infections/pathology , Retroviridae Infections/virology , Scleroderma, Localized/blood , Scleroderma, Localized/pathology , Scleroderma, Localized/virology , Ultraviolet Rays
3.
Nucleic Acids Res ; 43(15): 7414-31, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26160887

ABSTRACT

Ty1 Gag comprises the capsid of virus-like particles and provides nucleic acid chaperone (NAC) functions during retrotransposition in budding yeast. A subgenomic Ty1 mRNA encodes a truncated Gag protein (p22) that is cleaved by Ty1 protease to form p18. p22/p18 strongly inhibits transposition and can be considered an element-encoded restriction factor. Here, we show that only p22 and its short derivatives restrict Ty1 mobility whereas other regions of GAG inhibit mobility weakly if at all. Mutational analyses suggest that p22/p18 is synthesized from either of two closely spaced AUG codons. Interestingly, AUG1p18 and AUG2p18 proteins display different properties, even though both contain a region crucial for RNA binding and NAC activity. AUG1p18 shows highly reduced NAC activity but specific binding to Ty1 RNA, whereas AUG2p18 shows the converse behavior. p22/p18 affects RNA encapsidation and a mutant derivative defective for RNA binding inhibits the RNA chaperone activity of the C-terminal region (CTR) of Gag-p45. Moreover, affinity pulldowns show that p18 and the CTR interact. These results support the idea that one aspect of Ty1 restriction involves inhibition of Gag-p45 NAC functions by p22/p18-Gag interactions.


Subject(s)
Gene Products, gag/metabolism , Retroelements , Codon, Initiator , DNA, Viral/metabolism , Dimerization , Gene Products, gag/biosynthesis , Gene Products, gag/chemistry , Gene Products, gag/genetics , HIV-1/genetics , Protein Binding , Protein Biosynthesis , RNA/metabolism , RNA Caps/metabolism , RNA, Transfer, Met/metabolism , Saccharomyces/genetics
4.
Xenotransplantation ; 22(2): 144-50, 2015.
Article in English | MEDLINE | ID: mdl-25716804

ABSTRACT

BACKGROUND: Xenotransplantation represents one of alternative candidates for allotransplantation due to the chronic shortage of suitable human tissues; however, many obstacles remain. Expression and release of endogenous retroviral antigens by porcine cells after transplantation may evoke adverse immune responses in human subjects. Here, we examined whether human herpesvirus 1 (HHV-1) could induce the production of porcine endogenous retrovirus (PERV) antigens in porcine peripheral blood mononuclear cells (PBMCs). METHODS: Porcine PBMCs were infected with HHV-1 and examined for the production of PERV Gag protein and HHV-1 using antigen-capture ELISA and quantitative real-time polymerase chain reaction (PCR), respectively. RESULTS: HHV-1 infection resulted in a 1.7- to 33.2-fold induction of PERV Gag relative to mock infection controls, compared to a 2.9- to 12.9-fold induction following treatment with PMA. Expression of PERV Gag was detected in porcine PBMCs and PK-15 cells after HHV-1 infection by double immunofluorescence staining of PERV Gag and HHV-1 antigen. The viability of HHV-1-infected porcine PBMCs was significantly lower than that of mock-infected cells. The HHV-1 level in the culture supernatant increased 5.2-fold relative to controls 24-h post-infection, indicative of active replication within these cells; decreased levels of HHV-1 were detected 72-h post-infection. CONCLUSIONS: These results suggest that HHV-1 may be capable of infecting transplanted porcine cells, resulting in strong direct induction of PERV antigen.


Subject(s)
Antigens, Viral/biosynthesis , Endogenous Retroviruses/immunology , Herpesvirus 1, Human/pathogenicity , Heterografts/virology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Swine/virology , Animals , Cell Line , Coinfection/immunology , Coinfection/virology , Endogenous Retroviruses/pathogenicity , Enzyme-Linked Immunosorbent Assay/methods , Gene Products, gag/biosynthesis , Gene Products, gag/immunology , HEK293 Cells , Humans , Swine, Miniature , Transplantation, Heterologous/adverse effects
5.
J Gen Virol ; 95(Pt 9): 2050-2059, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24854000

ABSTRACT

The Gag polyprotein of feline immunodeficiency virus (FIV) assembles at the plasma membrane of the infected cells. We aimed to identify the FIV Gag domains that interact and promote Gag multimerization. To do this we generated a series of Gag subdomains and tested their ability to associate with full-length Gag and be recruited into extracellular virus-like particles (VLPs). Removal of 37 residues from the C-terminus of FIV Gag and deletion of the N-terminal and central regions of the nucleocapsid (NC) domain attenuated but did not abrogate association with wild-type Gag, whereas a Gag mutant protein encompassing the matrix (MA) and capsid (CA) domains interacted poorly with full-length Gag. Association with wild-type Gag was abolished by deleting most of the NC together with the N-terminal 40 residues of the MA, which most likely reflects the inability of this Gag mutant to bind RNA. Notably, the CA-NC Gag subdomain both associated with wild-type Gag and was recruited into particles in a proportion close to 50 % of the total Gag-related protein mass of VLPs. Moreover, both a Gag protein lacking the C-terminal p2 peptide and a nonmyristoylated version of the polyprotein exhibited a transdominant-negative effect on the assembly of wild-type Gag. Analysis of Gag mutants carrying internal deletions within the CA revealed that the N-terminal and the C-terminal domains of the CA are necessary for Gag assembly. Our results demonstrate that the FIV CA-NC region constitutes the principal self-interaction domain of Gag and that the RNA-binding capacity of Gag is necessary for its multimerization.


Subject(s)
Gene Products, gag/genetics , Immunodeficiency Virus, Feline/genetics , Protein Multimerization/genetics , Amino Acid Sequence , Animals , Binding Sites/genetics , COS Cells , Capsid/metabolism , Capsid Proteins/genetics , Cell Line , Cell Membrane/virology , Chlorocebus aethiops , Gene Products, gag/biosynthesis , Gene Products, gag/metabolism , Immunodeficiency Virus, Feline/pathogenicity , Molecular Sequence Data , Nucleocapsid/genetics , Protein Binding/genetics , Protein Structure, Tertiary/genetics , RNA-Binding Proteins/genetics , Rats , Sequence Alignment , Vaccinia virus/genetics , Vaccinia virus/pathogenicity , Viral Matrix Proteins/genetics , Virus Assembly/genetics
6.
J Virol ; 86(21): 11581-94, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22896611

ABSTRACT

Programmed -1 ribosomal frameshifting is widely used in the expression of RNA virus replicases and represents a potential target for antiviral intervention. There is interest in determining the extent to which frameshifting efficiency can be modulated before virus replication is compromised, and we have addressed this question using the alpharetrovirus Rous sarcoma virus (RSV) as a model system. In RSV, frameshifting is essential in the production of the Gag-Pol polyprotein from the overlapping gag and pol coding sequences. The frameshift signal is composed of two elements, a heptanucleotide slippery sequence and, just downstream, a stimulatory RNA structure that has been proposed to be an RNA pseudoknot. Point mutations were introduced into the frameshift signal of an infectious RSV clone, and virus replication was monitored following transfection and subsequent infection of susceptible cells. The introduced mutations were designed to generate a range of frameshifting efficiencies, yet with minimal impact on encoded amino acids. Our results reveal that point mutations leading to a 3-fold decrease in frameshifting efficiency noticeably reduce virus replication and that further reduction is severely inhibitory. In contrast, a 3-fold stimulation of frameshifting is well tolerated. These observations suggest that small-molecule inhibitors of frameshifting are likely to have potential as agents for antiviral intervention. During the course of this work, we were able to confirm, for the first time in vivo, that the RSV stimulatory RNA is indeed an RNA pseudoknot but that the pseudoknot per se is not absolutely required for virus viability.


Subject(s)
Frameshifting, Ribosomal , Rous sarcoma virus/physiology , Virus Replication , Base Sequence , Gene Products, gag/biosynthesis , Gene Products, pol/biosynthesis , Nucleic Acid Conformation , Point Mutation , RNA, Viral/chemistry , RNA, Viral/genetics , Rous sarcoma virus/genetics
7.
Virus Genes ; 47(2): 219-27, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23813246

ABSTRACT

Caffeine and aspirin have been suggested to be involved in neurologic diseases, such as schizophrenia, and previous data have revealed that abnormal expression of HERV-W elements may be an important factor in the etiopathogenesis of those diseases. In this article, we reported that caffeine and aspirin contributed to the expression of HERV-W env and gag in Human SH-SY5Y neuroblastoma cells. Semi-quantitative RT-PCR and quantitative Real-time PCR were used to detect the mRNA of HERV-W env and gag in cells exposed to caffeine or aspirin. Western blotting was used to detect the protein of HERV-W env. Luciferase activity assay was employed to detect the activity of HERV-W env promoter. It was found that both caffeine and aspirin could increase the expression of HERV-W env and gag in human SH-SY5Y neuroblastoma cells. Caffeine could activate the HERV-W env promoter, while aspirin could not. With previous studies we can conjecture that HERVs might play a bridging role between environmental factors, such as drugs and neurologic diseases.


Subject(s)
Aspirin/metabolism , Caffeine/metabolism , Endogenous Retroviruses/drug effects , Transcriptional Activation/drug effects , Blotting, Western , Cell Line, Tumor , Gene Expression Profiling , Gene Products, env/biosynthesis , Gene Products, env/genetics , Gene Products, gag/biosynthesis , Gene Products, gag/genetics , Genes, Reporter , Humans , Luciferases/analysis , Luciferases/genetics , Neurons/drug effects , Neurons/virology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction
8.
Proc Natl Acad Sci U S A ; 107(17): 7805-10, 2010 Apr 27.
Article in English | MEDLINE | ID: mdl-20385817

ABSTRACT

Retroviral particles assemble a few thousand units of the Gag polyproteins. Proteolytic cleavage mediated by the retroviral protease forms the bioactive retroviral protein subunits before cell entry. We hypothesized that this process could be exploited for targeted, transient, and dose-controlled transduction of nonretroviral proteins into cultured cells. We demonstrate that gammaretroviral particles tolerate the incorporation of foreign protein at several positions of their Gag or Gag-Pol precursors. Receptor-mediated and thus potentially cell-specific uptake of engineered particles occurred within minutes after cell contact. Dose and kinetics of nonretroviral protein delivery were dependent upon the location within the polyprotein precursor. Proteins containing nuclear localization signals were incorporated into retroviral particles, and the proteins of interest were released from the precursor by the retroviral protease, recognizing engineered target sites. In contrast to integration-defective lentiviral vectors, protein transduction by retroviral polyprotein precursors was completely transient, as protein transducing retrovirus-like particles could be produced that did not transduce genes into target cells. Alternatively, bifunctional protein-delivering particle preparations were generated that maintained their ability to serve as vectors for retroviral transgenes. We show the potential of this approach for targeted genome engineering of induced pluripotent stem cells by delivering the site-specific DNA recombinase, Flp. Protein transduction of Flp after proteolytic release from the matrix position of Gag allowed excision of a lentivirally transduced cassette that concomitantly expresses the canonical reprogramming transcription factors (Oct4, Klf4, Sox2, c-Myc) and a fluorescent marker gene, thus generating induced pluripotent stem cells that are free of lentivirally transduced reprogramming genes.


Subject(s)
Gene Products, gag/biosynthesis , Leukemia Virus, Murine/metabolism , Transduction, Genetic/methods , Virion/metabolism , Virus Internalization , Gene Products, gag/genetics , Genetic Engineering/methods , Green Fluorescent Proteins/metabolism , Kinetics , Leukemia Virus, Murine/genetics , Nuclear Localization Signals/metabolism , Peptide Hydrolases/metabolism , Virion/genetics
9.
Retrovirology ; 9: 8, 2012 Jan 25.
Article in English | MEDLINE | ID: mdl-22277305

ABSTRACT

BACKGROUND: Retroviral Gag proteins are encoded in introns and, because of this localization, they are subject to the default pathways of pre-mRNA splicing. Retroviruses regulate splicing and translation through a variety of intertwined mechanisms, including 5'- post-transcriptional control elements, 3'- constitutive transport elements, and viral protein RNA interactions that couple unspliced and singly spliced mRNAs to transport machinery. Sequences within the gag gene termed inhibitory or instability sequences also appear to affect viral mRNA stability and translation, and the action of these sequences can be countered by silent mutation or the presence of RNA interaction proteins like HIV-1 Rev. Here, we explored the requirements for mouse mammary tumor virus (MMTV) Gag expression using a combination of in vivo and in vitro expression systems. RESULTS: We show that MMTV gag alleles are inhibited for translation despite possessing a functional open reading frame (ORF). The block to expression was post-transcriptional and targeted the mRNA but was not a function of mRNA transport or stability. Using bicistronic reporters, we show that inhibition of gag expression imparted a block to both cap-dependent and cap-independent translation onto the mRNA. Direct introduction of in vitro synthesized gag mRNA resulted in translation, implying a nuclear role in inhibition of expression. The inhibition of expression was overcome by intact proviral expression or by flanking gag with splice sites combined with a functional Rem-Rem response element (RmRE) interaction. CONCLUSIONS: Expression of MMTV Gag requires nuclear interactions involving the viral Rem protein, its cognate binding target the RmRE, and surprisingly, both a splice donor and acceptor sequence to achieve appropriate signals for translation of the mRNA in the cytoplasm.


Subject(s)
Gene Expression Regulation, Viral , Gene Products, gag/biosynthesis , Mammary Tumor Virus, Mouse/physiology , Protein Biosynthesis , Animals , Cell Line , Gene Expression , Humans , Mammary Tumor Virus, Mouse/genetics , RNA Splicing
10.
J Virol ; 85(7): 3179-86, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21270144

ABSTRACT

Xenotropic murine leukemia virus-related virus (XMRV) is a gammaretrovirus linked to prostate carcinoma and chronic fatigue syndrome. Here we report that NF-κB activation can markedly increase XMRV production. The inflammatory cytokine tumor necrosis factor alpha (TNF-α), which activates NF-κB, significantly augmented viral Gag protein production in XMRV-infected cells. Reporter assays showed that TNF-α and Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1), an intrinsic NF-κB activator, increased long terminal repeat (LTR)-dependent XMRV transcription. We identified two NF-κB binding sites (designated κB-1 and κB-2) in the LTR U3 region of XMRV and demonstrated that both sites bind to the NF-κB component p65/RelA. Mutation of the κB-1 site, but not the κB-2 site, impaired responsiveness to TNF-α and LMP1 in reporter assays. A mutant XMRV with a mutation at the κB-1 site replicated significantly less efficiently than the wild-type XMRV in the prostate carcinoma LNCaP, DU145, and PC-3 cell lines, HEK293 cells, the EBV-immortalized cell line IB4, and the Burkitt's lymphoma cell line BJAB. These results demonstrate that TNF-α and EBV LMP1 enhance XMRV replication in prostate carcinoma and B-lineage cells through the κB-1 site in the XMRV LTR, suggesting that inflammation, EBV infection, and other conditions leading to NF-κB activation may promote XMRV spread in humans.


Subject(s)
B-Lymphocytes/virology , Carcinoma/virology , NF-kappa B/metabolism , Prostatic Neoplasms/virology , Transcription, Genetic , Virus Replication , Xenotropic murine leukemia virus-related virus/physiology , Binding Sites , Cell Line, Tumor , Gene Products, gag/biosynthesis , Humans , Male , Protein Binding , Terminal Repeat Sequences/genetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
PLoS Pathog ; 6(1): e1000748, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-20126442

ABSTRACT

The CD8+ T-cell is a key mediator of antiviral immunity, potentially contributing to control of pathogenic lentiviral infection through both innate and adaptive mechanisms. We studied viral dynamics during antiretroviral treatment of simian immunodeficiency virus (SIV) infected rhesus macaques following CD8+ T-cell depletion to test the importance of adaptive cytotoxic effects in clearance of cells productively infected with SIV. As previously described, plasma viral load (VL) increased following CD8+ T-cell depletion and was proportional to the magnitude of CD8+ T-cell depletion in the GALT, confirming a direct relationship between CD8+ T-cell loss and viral replication. Surprisingly, first phase plasma virus decay following administration of antiretroviral drugs was not slower in CD8+ T-cell depleted animals compared with controls indicating that the short lifespan of the average productively infected cell is not a reflection of cytotoxic T-lymphocyte (CTL) killing. Our findings support a dominant role for non-cytotoxic effects of CD8+ T-cells on control of pathogenic lentiviral infection and suggest that cytotoxic effects, if present, are limited to early, pre-productive stages of the viral life cycle. These observations have important implications for future strategies to augment immune control of HIV.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Viremia/immunology , Animals , Anti-Retroviral Agents/pharmacology , Gene Expression , Gene Products, gag/biosynthesis , Gene Products, gag/immunology , Gene Products, nef/biosynthesis , Gene Products, nef/immunology , Macaca mulatta , Models, Theoretical , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Viral Load/drug effects
12.
Proc Natl Acad Sci U S A ; 106(2): 605-10, 2009 Jan 13.
Article in English | MEDLINE | ID: mdl-19122141

ABSTRACT

The RNA silencing pathway is an intracellular innate response to virus infections and retro-transposons. Many plant viruses counter this host restriction by RNA silencing suppressor (RSS) activity of a double-stranded RNA-binding protein, e.g., tomato bushy stunt virus P19. Here, we demonstrate P19 and HIV-1 Tat function across the plant and animal kingdoms and suppress a common step in RNA silencing that is downstream of small RNA maturation. Our experiments reveal that RNA silencing in HIV-1 infected human cells severely attenuates the translational output of the unspliced HIV-1 gag mRNA, and possibly all HIV-1 transcripts. The attenuation in gag mRNA translation is exacerbated by K51A substitution in the Tat double-stranded RNA-binding domain. Tat, plant virus RSS, or Dicer downregulation rescues robust gag translation and bolsters HIV-1 virion production. The reversal of HIV-1 translation repression by plant RSS supports the recent finding in Arabidopsis that plant miRNAs operate by translational inhibition. Our results identify common features between RNA silencing suppression of plant and animal viruses. We suggest that RNA silencing-mediated translation repression plays a strategic role in determining the viral set-point in a newly HIV-1-infected patient.


Subject(s)
HIV-1/pathogenicity , Immunity, Innate , RNA Interference/immunology , Virus Replication , tat Gene Products, Human Immunodeficiency Virus/physiology , Cell Line , Gene Products, gag/biosynthesis , HIV Infections , HIV-1/genetics , Humans , Plant Viruses/genetics , Plant Viruses/pathogenicity , Protein Biosynthesis , RNA, Viral , Viral Proteins/physiology
13.
RNA Biol ; 8(2): 343-53, 2011.
Article in English | MEDLINE | ID: mdl-21422817

ABSTRACT

RNA-based compounds are promising agents to inactivate viruses. New specific hepatitis delta virus (HDV)-derived ribozymes are natural molecules that can be engineered to specifically target a viral RNA. We have designed specific on-off adaptor (SOFA)-HDV ribozymes targeting the tat and rev sequences of the human immunodeficiency virus type 1 (HIV-1) RNA. We show that the SOFA-HDV ribozymes cleave their RNA target in vitro. They inhibit the Tat-mediated transactivation of HIV-1 from 62% to 86% in different assays. In vivo, the amount of HIV RNA was decreased by 60 and 86% with two distinct ribozymes, which indicates that the inhibition of HIV production is directly correlated to the decline in spliced and unspliced viral RNAs. These SOFAHDV- ribozymes inhibited the expression and the viral production of four HIV-1 strains, indicating an extended potential to act on multiple HIV variants. In HEK 293T and HeLa cells transfected with pNL4-3 and the SOFA-HDV-ribozymes, the reduced RNA levels consequently decreased the Gag protein expression in the cell and virus production in the supernatant. When transfected before HIV-1 infection, the ribozymes prevented the incoming virus from being expressed. The ribozymes inhibited HIV production up to 90% when transfected in combination with the HIV protease inhibitor Atazanavir. Our results strongly suggest that SOFA-HDV ribozymes have a great potential to target HIV-1 and to be used as therapeutic agents in combination therapy.


Subject(s)
HIV-1/enzymology , RNA, Catalytic/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Virus Replication , Atazanavir Sulfate , Base Sequence , Gene Products, gag/biosynthesis , Gene Products, gag/genetics , Gene Products, rev/genetics , Gene Products, rev/metabolism , HEK293 Cells , HIV Infections/genetics , HIV-1/genetics , HeLa Cells , Hepatitis Delta Virus/enzymology , Hepatitis Delta Virus/genetics , Humans , Oligopeptides/pharmacology , Pyridines/pharmacology , RNA Splicing , RNA, Catalytic/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism
14.
Nucleic Acids Res ; 37(17): 5838-47, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19638424

ABSTRACT

The prototype foamy virus (PFV) is a nonpathogenic retrovirus that shows promise as a vector for gene transfer. The PFV (pre)genomic RNA starts with a long complex leader that can be folded into an elongated hairpin, suggesting an alternative strategy to cap-dependent linear scanning for translation initiation of the downstream GAG open reading frame (ORF). We found that the PFV leader carries several short ORFs (sORFs), with the three 5'-proximal sORFs located upstream of a structural element. Scanning-inhibitory hairpin insertion analysis suggested a ribosomal shunt mechanism, whereby ribosomes start scanning at the leader 5'-end and initiate at the downstream ORF via bypass of the central leader regions, which are inhibitory for scanning. We show that the efficiency of shunting depends strongly on the stability of the structural element located downstream of either sORFs A/A' or sORF B, and on the translation event at the corresponding 5'-proximal sORF. The PFV shunting strategy mirrors that of Cauliflower mosaic virus in plants; however, in mammals shunting can operate in the presence of a less stable structural element, although it is greatly improved by increasing the number of base pairings. At least one shunt configuration was found in primate FV (pre)genomic RNAs.


Subject(s)
5' Untranslated Regions , Peptide Chain Initiation, Translational , RNA, Viral/chemistry , Spumavirus/genetics , Animals , Cell Line , Gene Products, gag/biosynthesis , Gene Products, gag/genetics , Open Reading Frames , Ribosomes/metabolism
15.
J Exp Med ; 184(6): 2433-8, 1996 Dec 01.
Article in English | MEDLINE | ID: mdl-8976200

ABSTRACT

HIV-1 actively replicates in dendritic cell (DC)-T cell cocultures, but it has been difficult to demonstrate substantial infection of purified mature DCs. We now find that HIV-1 begins reverse transcription much more efficiently in DCs than T cells, even though T cells have higher levels of CD4 and gp120 binding. DCs isolated from skin or from blood precursors behave similarly. Several M-tropic strains and the T-tropic strain IIIB enter DCs efficiently, as assessed by the progressive formation of the early products of reverse transcription after a 90-min virus pulse at 37 degrees C. However, few late gag-containing sequences are detected, so that active viral replication does not occur. The formation of these early transcripts seems to follow entry of HIV-1, rather than binding of virions that contain viral DNA. Early transcripts are scarce if DCs are exposed to virus on ice for 4 h, or for 90 min at 37 degrees C, conditions which allow virus binding. Also the early transcripts once formed are insensitive to trypsin. The entry of a M-tropic isolates is blocked by the chemokine RANTES, and the entry of IIIB by SDF-1. RANTES interacts with CCR5 and SDF-1 with CXCR4 receptors. Entry of M-tropic but not T-tropic virus is ablated in DCs from individuals who lack a functional CCR5 receptor. DCs express more CCR5 and CXCR4 mRNA than T cells. Therefore, while HIV-1 does not replicate efficiently in mature DCs, viral entry can be active and can be blocked by chemokines that act on known receptors for M- and T-tropic virus.


Subject(s)
Dendritic Cells/virology , HIV-1/physiology , Receptors, Cytokine/immunology , T-Lymphocytes/virology , Virus Replication , Cells, Cultured , Chemokine CCL5/pharmacology , Coculture Techniques , Dendritic Cells/immunology , Gene Products, gag/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , HIV-1/drug effects , HIV-1/immunology , Humans , Interleukin-4/pharmacology , Polymerase Chain Reaction , Skin/immunology , T-Lymphocytes/immunology , Transcription, Genetic , Virion/immunology , Virion/physiology
16.
J Virol ; 83(23): 12483-98, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19776124

ABSTRACT

Retroviruses express Gag and Pol proteins by translation of unspliced genome-length viral RNA. For some retroviruses, transport of unspliced viral RNA to the cytoplasm is mediated by small regulatory proteins such as human immunodeficiency virus Rev, while other retroviruses contain constitutive transport elements in their RNAs that allow transport without splicing. In this study, we found that the betaretrovirus Jaagsiekte sheep retrovirus (JSRV) encodes within the env gene a trans-acting factor (Rej) necessary for the synthesis of Gag protein from unspliced viral RNA. Deletion of env sequences from a JSRV proviral expression plasmid (pTN3) abolished its ability to produce Gag polyprotein in transfected 293T cells, and Gag synthesis could be restored by cotransfection of an env expression plasmid (DeltaGP). Deletion analysis localized the complementing activity (Rej) to the putative Env signal peptide, and a signal peptide expression construct showed Rej activity. Two other betaretroviruses, mouse mammary tumor virus (MMTV) and human endogenous retrovirus type K, encode analogous factors (Rem and Rec, respectively) that are encoded from doubly spliced env mRNAs. Reverse transcriptase-PCR cloning and sequencing identified alternate internal splicing events in the 5' end of JSRV env that could signify analogous doubly spliced Rej mRNAs, and cDNA clones expressing two of them also showed Rej activity. The predicted Rej proteins contain motifs similar to those found in MMTV Rem and other analogous retroviral regulatory proteins. Interestingly, in most cell lines, JSRV expression plasmids with Rej deleted showed normal transport of unspliced JSRV RNA to the cytoplasm; however, in 293T cells Rej modestly enhanced export of unspliced viral RNA (2.8-fold). Metabolic labeling experiments with [(35)S]methionine indicated that JSRV Rej is required for the synthesis of viral Gag polyprotein. Thus, in most cell lines, the predominant function of Rej is to facilitate translation of unspliced viral mRNA.


Subject(s)
Carrier Proteins/physiology , Gene Products, gag/biosynthesis , Jaagsiekte sheep retrovirus/physiology , Viral Envelope Proteins/physiology , Virus Replication , Animals , Cell Line , Endogenous Retroviruses/genetics , Humans , Mammary Tumor Virus, Mouse/genetics , Molecular Sequence Data , Proviruses/genetics , Sequence Analysis, DNA , Sequence Deletion , Sequence Homology
17.
J Virol ; 83(23): 12499-511, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19776134

ABSTRACT

Jaagsiekte sheep retrovirus (JSRV) is a simple betaretrovirus causing a contagious lung cancer of sheep. JSRV encodes unspliced and spliced viral RNAs, among which unspliced RNA encodes Gag and Pol proteins and a singly spliced mRNA encodes Env protein. In another study we found that JSRV encodes a regulatory protein, Rej, that is responsible for synthesis of Gag polyprotein from unspliced viral RNA. Rej is encoded in the 5' end of env, and it enhances nuclear export or accumulation of cytoplasmic unspliced viral RNA in 293T cells but not in most other cell lines (A. Hofacre, T. Nitta, and H. Fan, J. Virol. 83:12483-12498, 2009). In this study, we found that mutations in the 3' end of env in the context of a cytomegalovirus-driven full-length JSRV expression construct abolished Gag protein synthesis and released viruses in 293T cells. These mutants also showed deficits in accumulation of unspliced viral RNA in the cytoplasm. These mutants defined a Rej-responsive element (RejRE). Inhibition of CRM1 but not Tap function prevented nuclear export/accumulation of cytoplasmic unspliced RNA in 293T cells, similarly to other complex retroviruses that express analogous regulator proteins (e.g., human immunodeficiency virus Rev). Structural modeling of the RejRE with Zuker M-fold indicated a region with a predicted stable secondary structure. Mutational analysis in this region indicated the importance of both secondary structures and primary nucleotide sequences in a central stem-bulge-stem structure. In contrast to 293T cells, mutations in the RejRE did not affect the levels of cytoplasmic unspliced RNA in 293 cells, although the unspliced RNA showed partial degradation, perhaps due to lack of translation. RejRE-containing RNA relocalized Rej protein from the nucleus to the cytoplasm in 293 and rat 208F cells, suggesting binding of Rej to the RejRE.


Subject(s)
Carrier Proteins/metabolism , Gene Products, gag/biosynthesis , Jaagsiekte sheep retrovirus/physiology , RNA, Viral/genetics , RNA, Viral/metabolism , Response Elements , Viral Envelope Proteins/metabolism , Virus Replication , Animals , Humans , Jaagsiekte sheep retrovirus/genetics , Mutagenesis, Site-Directed , Protein Structure, Secondary , Rats
18.
Clin Exp Immunol ; 160(3): 340-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20345981

ABSTRACT

Human endogenous retroviruses (HERVs) are remnants of ancient retroviral infections within the human genome. These molecular fossils draw parallels with present-day exogenous retroviruses and have been linked previously with immunopathology within rheumatoid arthritis (RA). Mechanisms of pathogenesis for HERV-K in RA such as molecular mimicry were investigated. To clarify a role for HERVs in RA, potential autoantigens implicated in autoimmunity were scanned for sequence identity with retroviral epitopes. Short retroviral peptides modelling shared epitopes were synthesized, to survey anti-serum of RA patients and disease controls. A novel real-time polymerase chain reaction (PCR) assay was also developed to quantify accurately levels of HERV-K (HML-2) gag expression, relative to normalized housekeeping gene expression. Both serological and molecular assays showed significant increases in HERV-K (HML-2) gag activity in RA patients, compared to disease controls. The real-time PCR assay identified significant up-regulation in HERV-K mRNA levels in RA patients compared to inflammatory and healthy controls. Exogenous viral protein expression and proinflammatory cytokines were also shown to exert modulatory effects over HERV-K (HML-2) transcription. From our data, it can be concluded that RA patients exhibited significantly elevated levels of HERV-K (HML-2) gag activity compared to controls. Additional factors influencing HERV activity within the synovium were also identified. The significant variation in RA patients, both serologically and transcriptionally, may be an indication that RA is an umbrella term for a number of separate disease entities, of which particular HERV polymorphisms may play a role in development.


Subject(s)
Arthritis, Rheumatoid/metabolism , Autoantigens/metabolism , Endogenous Retroviruses/metabolism , Gene Expression Regulation, Viral/immunology , Gene Products, gag/biosynthesis , Molecular Mimicry , Peptides/metabolism , Adult , Aged , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/virology , Autoantigens/immunology , Endogenous Retroviruses/immunology , Epitopes/immunology , Epitopes/metabolism , Female , Gene Products, gag/immunology , Humans , Male , Middle Aged , Peptides/immunology , Polymorphism, Genetic/immunology , RNA, Messenger/biosynthesis , RNA, Messenger/immunology , RNA, Viral/biosynthesis , RNA, Viral/immunology , Synovial Membrane/immunology , Synovial Membrane/metabolism , Synovial Membrane/virology , Transcription, Genetic/immunology
19.
J Virol ; 82(10): 5031-42, 2008 May.
Article in English | MEDLINE | ID: mdl-18337567

ABSTRACT

The brains of individuals with lentiviral-associated encephalitis contain an abundance of infected and activated macrophages. It has been hypothesized that encephalitis develops when increased numbers of infected monocytes traffic into the central nervous system (CNS) during the end stages of immunosuppression. The relationships between the infection of brain and systemic macrophages and circulating monocytes and the development of lentiviral encephalitis are unknown. We longitudinally examined the extent of monocyte/macrophage infection in blood and lymph nodes of pigtailed macaques that did or did not develop simian immunodeficiency virus encephalitis (SIVE). Compared to levels in macaques that did not develop SIVE, more ex vivo virus production was detected from monocyte-derived macrophages and nonadherent peripheral blood mononuclear cells (PBMCs) from macaques that did develop SIVE. Prior to death, there was an increase in the number of circulating PBMCs following a rise in cerebrospinal fluid viral load in macaques that did develop SIVE but not in nonencephalitic macaques. At necropsy, macaques with SIVE had more infected macrophages in peripheral organs, with the exception of lymph nodes. T cells and NK cells with cytotoxic potential were more abundant in brains with encephalitis; however, T-cell and NK-cell infiltration in SIVE and human immunodeficiency virus encephalitis was more modest than that observed in classical acute herpes simplex virus encephalitis. These findings support the hypothesis that inherent differences in host systemic and CNS monocyte/macrophage viral production are associated with the development of encephalitis.


Subject(s)
Encephalitis/immunology , Encephalitis/virology , Macrophages/virology , Simian Acquired Immunodeficiency Syndrome/complications , Simian Acquired Immunodeficiency Syndrome/immunology , Animals , Brain/immunology , Brain/pathology , CD4 Lymphocyte Count , Cerebrospinal Fluid/virology , Gene Products, gag/biosynthesis , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/virology , Longitudinal Studies , Lymph Nodes/immunology , Lymph Nodes/virology , Macaca nemestrina , RNA, Viral/cerebrospinal fluid , Simian Immunodeficiency Virus/growth & development , T-Lymphocytes/immunology , Viral Load
20.
J Cell Biol ; 135(6 Pt 2): 1841-52, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8991095

ABSTRACT

Retrovirus Moloney murine leukemia virus (M-MuLV) matures by budding at the cell surface. Central to the budding process is the myristoylated viral core protein precursor Gag which, even in the absence of all other viral components, is capable of associating with the cytoplasmic leaflet of the plasma membrane and assembling into extracellular virus-like particles. In this paper we have used heterologous, Semliki Forest virus-driven, expression of M-MuLV Gag to study the mechanism by which this protein is targeted to the cell surface. In pulse-chase experiments, BFA, monensin, and 20 degrees C block did not affect incorporation of Gag into extracellular particles thereby indicating that the secretory pathway is not involved in targeting of Gag to the cell surface. Subcellular fractionation studies demonstrated that newly synthesized Gag became rapidly and efficiently associated with membranes which had a density similar to that of plasma membrane-derived vesicles. Protease-protection studies confirmed that the Gag-containing membranes were of plasma membrane origin, since in crude cell homogenates, the bulk of newly synthesized Gag was protease-resistant as expected of a protein that binds to the cytoplasmic leaflet of the plasma membrane. Taken together these data indicate that targeting of M-MuLV Gag to the cell surface proceeds via direct insertion of the protein to the cytoplasmic side of the plasma membrane. Furthermore, since the membrane insertion reaction is highly efficient and specific, this suggests that the reaction is dependent on as-yet-unidentified cellular factors.


Subject(s)
Gene Products, gag/metabolism , Moloney murine leukemia virus/chemistry , Protein Precursors/metabolism , Virus Replication , Antiviral Agents/pharmacology , Biological Transport/drug effects , Biological Transport/physiology , Brefeldin A , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Cold Temperature , Cyclopentanes/pharmacology , Endopeptidases , Gene Expression Regulation, Viral/physiology , Gene Products, gag/biosynthesis , Gene Products, gag/genetics , Genome, Viral , Ionophores/pharmacology , Kinetics , Microscopy, Electron , Moloney murine leukemia virus/growth & development , Moloney murine leukemia virus/ultrastructure , Monensin/pharmacology , Protein Precursors/genetics , Recombinant Proteins/metabolism , Semliki forest virus/chemistry , Semliki forest virus/genetics , Subcellular Fractions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL