Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 342
Filtrar
1.
J Virol ; 95(15): e0017021, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-33980598

RESUMO

Murine leukemia virus (MLV) requires the infected cell to divide to access the nucleus to integrate into the host genome. It has been determined that MLV uses the microtubule and actin network to reach the nucleus at the early stages of infection. Several studies have shown that viruses use the dynein motor protein associated with microtubules for their displacement. We have previously reported that dynein light-chain roadblock type 2 (Dynlrb2) knockdown significantly decreases MLV infection compared to nonsilenced cells, suggesting a functional association between this dynein light chain and MLV preintegration complex (PIC). In this study, we aimed to determine if the dynein complex Dynlrb2 subunit plays an essential role in the retrograde transport of MLV. For this, an MLV mutant containing the green fluorescent protein (GFP) fused to the viral protein p12 was used to assay the PIC localization and speed in cells in which the expression of Dynlrb2 was modulated. We found a significant decrease in the arrival of MLV PIC to the nucleus and a reduced net speed of MLV PICs when Dynlrb2 was knocked down. In contrast, an increase in nuclear localization was observed when Dynlrb2 was overexpressed. Our results suggest that Dynlrb2 plays an essential role in MLV retrograde transport. IMPORTANCE Different viruses use different components of cytoplasmic dynein complex to traffic to their replication site. We have found that murine leukemia virus (MLV) depends on dynein light-chain Dynlrb2 for infection, retrograde traffic, and nuclear entry. Our study provides new information regarding the molecular requirements for retrograde transport of MLV preintegration complex and demonstrates the essential role of Dynlrb2 in MLV infection.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Dineínas do Citoplasma/genética , Dineínas/metabolismo , Vírus da Leucemia Murina/crescimento & desenvolvimento , Replicação Viral/genética , Células 3T3 , Transporte Ativo do Núcleo Celular/genética , Animais , Linhagem Celular , Núcleo Celular/virologia , Dineínas/genética , Produtos do Gene gag/genética , Células HEK293 , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Camundongos , Microtúbulos/metabolismo
2.
Arch Virol ; 165(5): 1089-1097, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32146506

RESUMO

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.


Assuntos
Vetores Genéticos , Vírus da Leucemia do Macaco Gibão/crescimento & desenvolvimento , Vírus da Leucemia Murina/crescimento & desenvolvimento , Vesiculovirus/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Replicação Viral , Animais , Linhagem Celular , Terapia Genética/métodos , Humanos , Vírus da Leucemia do Macaco Gibão/genética , Vírus da Leucemia Murina/genética , Camundongos , Neoplasias/terapia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Recombinação Genética
3.
J Virol Methods ; 274: 113714, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31412271

RESUMO

Hepatitis C pseudoparticles (HCVpp) are used to evaluate HCV cell entry while screening for neutralizing antibodies induced upon vaccination or while screening for new antiviral drugs. In this work we explore the stable production of HCVpp aiming to reduce the variability associated with transient productions. The performance of stably produced HCVpp was assessed by evaluating the influence of Human Serum and the impact of CD81 cellular expression on the infectivity of HCVpp. After evaluating the performance of stably produced HCVpp we studied the effect of co-expressing p7NS2 openreading frame (ORF) on HCVpp infectivity. Our data clearly shows an enhanced infectivity of HCVppp7NS2. Even though the exact mechanism was not completely elucidated, the enhanced infectivity of HCVppp7NS2 is neither a result of an increase production of virus particles nor a result from increased envelope density. The inhibitory effect of p7 inhibitory molecules such as rimantadine suggests a direct contribution of p7 ion channel for the enhanced infectivity of HCVppp7NS2 which is coherent with a pH-dependent cell entry mechanism. In conclusion, we report the establishment of a stable production system of HCVpp with enhanced infectivity through the overexpression of p7NS2 ORF contributing to improve HCV entry assessment assays widely used in antiviral drug discovery and vaccine development.


Assuntos
Expressão Gênica , Hepacivirus/fisiologia , Vírus da Leucemia Murina/crescimento & desenvolvimento , Proteínas não Estruturais Virais/biossíntese , Proteínas Virais/biossíntese , Cultura de Vírus/métodos , Internalização do Vírus , Linhagem Celular , Genes Reporter , Vetores Genéticos , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Hepacivirus/genética , Humanos , Vírus da Leucemia Murina/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Coloração e Rotulagem/métodos , Proteínas não Estruturais Virais/genética , Proteínas Virais/genética
4.
PLoS One ; 11(12): e0167293, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27936017

RESUMO

Prion diseases are infectious and fatal neurodegenerative diseases which require the cellular prion protein, PrPC, for development of diseases. The current study shows that the PrPC augments infectivity and plaque formation of a mouse endogenous retrovirus, MuLV. We have established four neuronal cell lines expressing mouse PrPC, PrP+/+; two express wild type PrPC (MoPrPwild) and the other two express mutant PrPC (MoPrPmut). Infection of neuronal cells from various PrP+/+ and PrP-/- (MoPrPKO) lines with MuLV yielded at least three times as many plaques in PrP+/+ than in PrP-/-. Furthermore, among the four PrP+/+ lines, one mutant line, P101L, had at least 2.5 times as many plaques as the other three PrP+/+ lines. Plaques in P101L were four times larger than those in other PrP+/+ lines. Colocalization of PrP and CAgag was seen in MuLV-infected PrP+/+ cells. In the PrP-MuLV interaction, the involvement of galectin-3 and -6 was observed by immunoprecipitation with antibody to PrPC. These results suggest that PrPC combined with galectin-3 and -6 can act as a receptor for MuLV. P101L, the disease form of mutant PrPC results suggest the genetic mutant form of PrPC may be more susceptible to viral infection.


Assuntos
Galectina 3/metabolismo , Galectinas/metabolismo , Vírus da Leucemia Murina/crescimento & desenvolvimento , Neurônios/metabolismo , Proteínas PrPC/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Astrócitos/metabolismo , Astrócitos/virologia , Western Blotting , Linhagem Celular , Células Cultivadas , Retrovirus Endógenos/crescimento & desenvolvimento , Retrovirus Endógenos/fisiologia , Galectina 3/genética , Galectinas/genética , Hipocampo/citologia , Hipocampo/virologia , Interações Hospedeiro-Patógeno , Vírus da Leucemia Murina/fisiologia , Camundongos Knockout , Microscopia Confocal , Neurônios/citologia , Neurônios/virologia , Proteínas PrPC/genética , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Biotechnol Prog ; 32(1): 89-97, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26488618

RESUMO

To ensure the viral safety of protein therapeutics made in mammalian cells, purification processes include dedicated viral clearance steps to remove or inactivate adventitious and endogenous viruses. One such dedicated step is low pH treatment, a robust and effective method commonly used in monoclonal antibody production to inactivate enveloped viruses. To characterize the operating space for low pH viral inactivation, we performed a statistically designed experiment evaluating the effect of pH, temperature, hold duration, acid type, and buffer concentration on inactivation of the retrovirus model, XMuLV. An additional single factor experiment was performed to study the effect of protein concentration. These data were used to generate predictive models of inactivation at each time point studied, which can be used to identify conditions for robust and effective XMuLV inactivation. At pH 3.6, XMuLV inactivation was rapid, robust, and relatively unaffected by the other factors studied, providing support for this as a generic viral inactivation condition for products that can tolerate this low pH. At pH 3.7 and 3.8, other factors besides pH affected XMuLV inactivation. By understanding the impact of each factor on inactivation, the factors can be manipulated within the operating space to ensure effective inactivation while achieving desired product quality goals.


Assuntos
Vírus da Leucemia Murina/crescimento & desenvolvimento , Proteínas do Envelope Viral/química , Inativação de Vírus , Animais , Anticorpos Monoclonais/imunologia , Biotecnologia , Concentração de Íons de Hidrogênio , Cinética , Vírus da Leucemia Murina/patogenicidade , Retroviridae/efeitos dos fármacos , Temperatura , Proteínas do Envelope Viral/imunologia
6.
Nature ; 491(7426): 774-8, 2012 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-23103862

RESUMO

The mammalian host has developed a long-standing symbiotic relationship with a considerable number of microbial species. These include the microbiota on environmental surfaces, such as the respiratory and gastrointestinal tracts, and also endogenous retroviruses (ERVs), comprising a substantial fraction of the mammalian genome. The long-term consequences for the host of interactions with these microbial species can range from mutualism to parasitism and are not always completely understood. The potential effect of one microbial symbiont on another is even less clear. Here we study the control of ERVs in the commonly used C57BL/6 (B6) mouse strain, which lacks endogenous murine leukaemia viruses (MLVs) able to replicate in murine cells. We demonstrate the spontaneous emergence of fully infectious ecotropic MLV in B6 mice with a range of distinct immune deficiencies affecting antibody production. These recombinant retroviruses establish infection of immunodeficient mouse colonies, and ultimately result in retrovirus-induced lymphomas. Notably, ERV activation in immunodeficient mice is prevented in husbandry conditions associated with reduced or absent intestinal microbiota. Our results shed light onto a previously unappreciated role for immunity in the control of ERVs and provide a potential mechanistic link between immune activation by microbial triggers and a range of pathologies associated with ERVs, including cancer.


Assuntos
Anticorpos Antivirais/biossíntese , Retrovirus Endógenos/fisiologia , Hospedeiro Imunocomprometido/imunologia , Ativação Viral , Criação de Animais Domésticos , Animais , Anticorpos Antivirais/imunologia , Transformação Celular Viral , Retrovirus Endógenos/genética , Retrovirus Endógenos/crescimento & desenvolvimento , Retrovirus Endógenos/imunologia , Feminino , Leucemia/virologia , Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/crescimento & desenvolvimento , Vírus da Leucemia Murina/imunologia , Vírus da Leucemia Murina/fisiologia , Linfoma/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/deficiência , Receptores de Antígenos de Linfócitos T/genética , Recombinação Genética , Viremia/imunologia , Viremia/virologia
7.
J Virol ; 86(13): 7241-8, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22514353

RESUMO

Previous studies indicate that mice infected with mixtures of mouse retroviruses (murine leukemia viruses [MuLVs]) exhibit dramatically altered pathology compared to mice infected with individual viruses of the mixture. Coinoculation of the ecotropic virus Friend MuLV (F-MuLV) with Fr98, a polytropic MuLV, induced a rapidly fatal neurological disease that was not observed in infections with either virus alone. The polytropic virus load in coinoculated mice was markedly enhanced, while the ecotropic F-MuLV load was unchanged. Furthermore, pseudotyping of the polytropic MuLV genome within ecotropic virions was nearly complete in coinoculated mice. In an effort to better understand these phenomena, we examined mixed retrovirus infections by utilizing in vitro cell lines. Similar to in vivo mixed infections, the polytropic MuLV genome was extensively pseudotyped within ecotropic virions; polytropic virus release was profoundly elevated in coinfected cells, and the ecotropic virus release was unchanged. A reduced level of polytropic SU protein on the surfaces of coinfected cells was observed and correlated with a reduced level of nonpseudotyped polytropic virion release. Marked amplification and pseudotyping of the polytropic MuLV were also observed in mixed Fr98-F-MuLV infections of cell lines derived from the central nervous system (CNS), the target for Fr98 pathogenesis. Additional experiments indicated that pseudotyping contributed to the elevated polytropic virus titer by increasing the efficiency of packaging and release of the polytropic genomes within ecotropic virions. Mixed infections are the rule rather than the exception in retroviral infection, and the ability to examine them in vitro should facilitate a more thorough understanding of retroviral interactions in general.


Assuntos
Coinfecção/virologia , Vírus da Leucemia Murina/crescimento & desenvolvimento , Vírus da Leucemia Murina/fisiologia , Liberação de Vírus , Animais , Linhagem Celular , Camundongos , Carga Viral
8.
Retrovirology ; 9: 17, 2012 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-22348230

RESUMO

BACKGROUND: Ankyrins are cellular mediators of a number of essential protein-protein interactions. Unlike intrabodies, ankyrins are composed of highly structured repeat modules characterized by disulfide bridge-independent folding. Artificial ankyrin molecules, designed to target viral components, might act as intracellular antiviral agents and contribute to the cellular immunity against viral pathogens such as HIV-1. RESULTS: A phage-displayed library of artificial ankyrins was constructed, and screened on a polyprotein made of the fused matrix and capsid domains (MA-CA) of the HIV-1 Gag precursor. An ankyrin with three modules named Ank(GAG)1D4 (16.5 kDa) was isolated. Ank(GAG)1D4 and MA-CA formed a protein complex with a stoichiometry of 1:1 and a dissociation constant of K(d) ~ 1 µM, and the Ank(GAG)1D4 binding site was mapped to the N-terminal domain of the CA, within residues 1-110. HIV-1 production in SupT1 cells stably expressing Ank(GAG)1D4 in both N-myristoylated and non-N-myristoylated versions was significantly reduced compared to control cells. Ank(GAG)1D4 expression also reduced the production of MLV, a phylogenetically distant retrovirus. The Ank(GAG)1D4-mediated antiviral effect on HIV-1 was found to occur at post-integration steps, but did not involve the Gag precursor processing or cellular trafficking. Our data suggested that the lower HIV-1 progeny yields resulted from the negative interference of Ank(GAG)1D4-CA with the Gag assembly and budding pathway. CONCLUSIONS: The resistance of Ank(GAG)1D4-expressing cells to HIV-1 suggested that the CA-targeted ankyrin Ank(GAG)1D4 could serve as a protein platform for the design of a novel class of intracellular inhibitors of HIV-1 assembly based on ankyrin-repeat modules.


Assuntos
Anquirinas/farmacologia , Fármacos Anti-HIV/farmacologia , HIV-1/efeitos dos fármacos , Produtos do Gene gag do Vírus da Imunodeficiência Humana/antagonistas & inibidores , Sequência de Aminoácidos , Linhagem Celular , HIV-1/crescimento & desenvolvimento , Humanos , Vírus da Leucemia Murina/efeitos dos fármacos , Vírus da Leucemia Murina/crescimento & desenvolvimento , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Mapeamento de Interação de Proteínas , Proteínas Recombinantes/farmacologia , Montagem de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
9.
Retrovirology ; 8: 53, 2011 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-21729311

RESUMO

BACKGROUND: Over the last several decades it has been noted, using a variety of different methods, that cells infected by a specific gammaretrovirus are resistant to infection by other retroviruses that employ the same receptor; a phenomenon termed receptor interference. Receptor masking is thought to provide an earlier means of blocking superinfection, whereas receptor down regulation is generally considered to occur in chronically infected cells. RESULTS: We used replication-competent GFP-expressing viruses containing either an amphotropic murine leukemia virus (A-MLV) or the gibbon ape leukemia virus (GALV) envelope. We also constructed similar viruses containing fluorescence-labeled Gag proteins for the detection of viral particles. Using this repertoire of reagents together with a wide range of antibodies, we were able to determine the presence and availability of viral receptors, and detect viral envelope proteins and particles presence on the cell surface of chronically infected cells. CONCLUSIONS: A-MLV or GALV receptors remain on the surface of chronically infected cells and are detectable by respective antibodies, indicating that these receptors are not downregulated in these infected cells as previously proposed. We were also able to detect viral envelope proteins on the infected cell surface and infected cells are unable to bind soluble A-MLV or GALV envelopes indicating that receptor binding sites are masked by endogenously expressed A-MLV or GALV viral envelope. However, receptor masking does not completely prevent A-MLV or GALV superinfection.


Assuntos
Interações Hospedeiro-Patógeno , Vírus da Leucemia do Macaco Gibão/fisiologia , Vírus da Leucemia Murina/fisiologia , Receptores Virais/biossíntese , Animais , Bovinos , Linhagem Celular , Cricetinae , Cricetulus , Regulação para Baixo , Genes Reporter , Humanos , Vírus da Leucemia do Macaco Gibão/crescimento & desenvolvimento , Vírus da Leucemia Murina/crescimento & desenvolvimento , Camundongos
10.
J Immunol ; 186(5): 3023-30, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21270402

RESUMO

Retroviruses need to bud from producer cells to spread infection. To facilitate its budding, some virus hijacks the multivesicular body (MVB) pathway that is normally used to cargo and degrade ubiquitylated cellular proteins, through interaction between the late domain of Gag polyproteins and the components of MVB machinery. In this study, we demonstrated that TANK-binding kinase 1 (TBK1) directly interacted with VPS37C, a subunit of endosomal sorting complex required for transport-I (ESCRT-I) in the MVB pathway, without affecting the ultrastructure or general function of MVB. Interestingly, overexpression of TBK1 attenuated, whereas short hairpin RNA interference of TBK1 enhanced HIV-1 pseudovirus release from Vero cells in type I IFN (IFN-I)-independent manner. Down-regulation of TBK1 by short hairpin RNA in TZM-bl cells also enhanced live HIV-1 NL4-3 or JR-CSF virus budding without involvement of IFN-I induction. Furthermore, infection of TBK1-deficient mouse embryonic fibroblast cells with a chimeric murine leukemia virus/p6, whose PPPY motif was replaced by PTAP motif of HIV-1, showed that lack of TBK1 significantly enhanced PTAP-dependent, but not PPPY-dependent retrovirus budding. Finally, phosphorylation of VPS37C by TBK1 might regulate the viral budding efficiency, because overexpression of the kinase-inactive mutant of TBK1 (TBK1-K38A) in Vero cells accelerated HIV-1 pseudovirus budding. Therefore, through tethering to VPS37C of the ESCRT-I complex, TBK1 controlled the speed of PTAP-dependent retroviral budding through phosphorylation of VPS37C, which would serve as a novel mechanism of host cell defense independent of IFN-I signaling.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Vírus da Leucemia Murina/imunologia , Proteínas Serina-Treonina Quinases/fisiologia , Replicação Viral/imunologia , Animais , Linhagem Celular , Chlorocebus aethiops , Células HEK293 , Infecções por HIV/imunologia , Infecções por HIV/metabolismo , HIV-1/imunologia , Humanos , Vírus da Leucemia Murina/crescimento & desenvolvimento , Camundongos , Camundongos Knockout , Corpos Multivesiculares/fisiologia , Corpos Multivesiculares/ultraestrutura , Corpos Multivesiculares/virologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/deficiência , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/metabolismo , Infecções por Retroviridae/virologia , Transdução de Sinais/imunologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/metabolismo , Células Vero
11.
J Immunol ; 185(6): 3305-12, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20693424

RESUMO

Indoleamine 2,3-dioxygenase, the L-tryptophan-degrading enzyme, plays a key role in the powerful immunomodulatory effects on several different types of cells. Because modulation of IDO activities after viral infection may have great impact on disease progression, we investigated the role of IDO following infection with LP-BM5 murine leukemia virus. We found suppressed BM5 provirus copies and increased type I IFNs in the spleen from IDO knockout (IDO(-/-)) and 1-methyl-D-L-tryptophan-treated mice compared with those from wild-type (WT) mice. Additionally, the number of plasmacytoid dendritic cells in IDO(-/-) mice was higher in the former than in the WT mice. In addition, neutralization of type I IFNs in IDO(-/-) mice resulted in an increase in LP-BM5 viral replication. Moreover, the survival rate of IDO(-/-) mice or 1-methyl-D-L-tryptophan-treated mice infected with LP-BM5 alone or with both Toxoplasma gondii and LP-BM5 was clearly greater than the survival rate of WT mice. To our knowledge, the present study is the first report to observe suppressed virus replication with upregulated type I IFN in IDO(-/-) mice, suggesting that modulation of the IDO pathway may be an effective strategy for treatment of virus infection.


Assuntos
Regulação para Baixo/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/deficiência , Interferon Tipo I/biossíntese , Vírus da Leucemia Murina/imunologia , Infecções por Retroviridae/enzimologia , Infecções por Retroviridae/prevenção & controle , Regulação para Cima/imunologia , Replicação Viral/imunologia , Imunidade Adaptativa/genética , Animais , Regulação para Baixo/genética , Imunidade Inata/genética , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Interferon Tipo I/fisiologia , Vírus da Leucemia Murina/crescimento & desenvolvimento , Leucemia Experimental/enzimologia , Leucemia Experimental/imunologia , Leucemia Experimental/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Retroviridae/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Infecções Tumorais por Vírus/enzimologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/prevenção & controle , Regulação para Cima/genética
12.
Retrovirology ; 6: 86, 2009 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-19772602

RESUMO

BACKGROUND: Contamination of vertebrate cell lines with animal retroviruses has been documented repeatedly before. Although such viral contaminants can be easily identified with high sensitivity by PCR, it is impossible to screen for all potential contaminants. Therefore, we explored two novel methods to identify viral contaminations in cell lines without prior knowledge of the kind of contaminant. RESULTS: The first hint for the presence of contaminating retroviruses in one of our cell lines was obtained by electron microscopy of exosome-like vesicles released from the supernatants of transfected 293T cells. Random amplification of particle associated RNAs (PAN-PCR) from supernatant of contaminated 293T cells and sequencing of the amplicons revealed several nucleotide sequences showing highest similarity to either murine leukemia virus (MuLV) or squirrel monkey retrovirus (SMRV). Subsequent mass spectrometry analysis confirmed our findings, since we could identify several peptide sequences originating from monkey and murine retroviral proteins. Quantitative PCRs were established for both viruses to test currently cultured cell lines as well as liquid nitrogen frozen cell stocks. Gene fragments for both viruses could be detected in a broad range of permissive cell lines from multiple species. Furthermore, experimental infections of cells negative for these viruses showed that both viruses replicate rapidly to high loads. We decided to further analyze the genomic sequence of the MuLV-like contaminant virus. Surprisingly it was neither identical to MuLV nor to the novel xenotropic MuLV related retrovirus (XMRV) but showed 99% identity to a synthetic retrovirus which was engineered in the 1980s. CONCLUSION: The high degree of nucleotide identity suggests unintended spread of a biosafety level 2 recombinant virus, which could also affect the risk assessment of gene-modified organisms released from contaminated cell cultures. The study further indicates that both mass spectrometry and PAN-PCR are powerful methods to identify viral contaminations in cell lines without prior knowledge of the kind of contaminant. Both methods might be useful tools for testing cell lines before using them for critical purposes.


Assuntos
Betaretrovirus/crescimento & desenvolvimento , Betaretrovirus/isolamento & purificação , Linhagem Celular/virologia , Vírus da Leucemia Murina/crescimento & desenvolvimento , Vírus da Leucemia Murina/isolamento & purificação , Contenção de Riscos Biológicos , Humanos , Espectrometria de Massas/métodos , Microscopia Eletrônica/métodos , Reação em Cadeia da Polimerase/métodos
13.
Cell Host Microbe ; 6(1): 68-80, 2009 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-19616766

RESUMO

Primate lentiviruses, including HIV-1, transduce terminally differentiated, nondividing myeloid cells; however, these cells are refractory to infection by gammaretroviruses such as murine leukemia virus (MLV). Here, we present evidence that a cellular restriction is the obstacle to transduction of macrophages by MLV. Neutralization of the restriction by Vpx, a primate lentiviral protein previously shown to protect primate lentiviruses from a macrophage restriction, rendered macrophages permissive to MLV infection. We further demonstrate that this restriction prevents transduction of quiescent monocytes by HIV-1. Monocyte-HeLa heterokaryons were resistant to HIV-1 infection, while heterokaryons formed between monocytes and HeLa cells expressing Vpx were permissive to HIV-1 infection. Encapsidation of Vpx within HIV-1 virions conferred the ability to infect quiescent monocytes. Collectively, our results indicate that the relative ability of lentiviruses and gammaretroviruses to transduce nondividing myeloid cells is dependent upon their ability to neutralize a cellular restriction.


Assuntos
HIV-1/crescimento & desenvolvimento , HIV-1/imunologia , Vírus da Leucemia Murina/crescimento & desenvolvimento , Vírus da Leucemia Murina/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Linhagem Celular , Células Cultivadas , Humanos , Macrófagos/virologia , Monócitos/virologia , Transdução Genética , Proteínas Virais Reguladoras e Acessórias/imunologia
14.
Nature ; 458(7242): 1201-4, 2009 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-19270682

RESUMO

Embryonic stem cells (ESCs) and other primitive stem cells of mice have been known for more than 30 years to potently block retrovirus replication. Infection of ESCs by the murine leukaemia viruses (MLVs) results in the normal establishment of integrated proviral DNA, but this DNA is then transcriptionally silenced, preventing further viral spread. The repression is largely mediated by trans-acting factors that recognize a conserved sequence element termed the primer binding site, an 18-base pair sequence complementary to the 3' end of a cellular transfer RNA. A specific tRNA is annealed to the primer binding site sequence of the viral genomic RNA, and is used to prime DNA synthesis. This same sequence in the context of the integrated proviral DNA is targeted for silencing in ESCs. We have recently shown that a large protein complex binding to the primer binding site in ESCs contains TRIM28 (refs 8, 9), a well-characterized transcriptional co-repressor. An important question remains as to the identity of the factor that directly recognizes integrated retroviral DNAs and recruits TRIM28 to mediate their specific silencing. Here we identify the zinc finger protein ZFP809 as the recognition molecule that bridges the integrated proviral DNA and TRIM28. We show that expression of ZFP809 is sufficient to render even differentiated cells highly resistant to MLV infection. Furthermore, we demonstrate that ZFP809 is able to potently block transcription from DNA constructs of human T-cell lymphotropic virus-1 (HTLV-1), which use the same primer tRNA. These results identify ZFP809 as a DNA-binding factor that specifically recognizes a large subset of mammalian retroviruses and retroelements, targeting them for transcriptional silencing. We propose that ZFP809 evolved as a stem-cell-specific retroviral restriction factor, and therefore constitutes a new component of the intrinsic immune system of stem cells.


Assuntos
DNA Viral/genética , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/virologia , Regulação Viral da Expressão Gênica , Inativação Gênica , Retroviridae/genética , Animais , Diferenciação Celular , Linhagem Celular , DNA Viral/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/isolamento & purificação , Vírus Linfotrópico T Tipo 1 Humano/genética , Vírus Linfotrópico T Tipo 1 Humano/crescimento & desenvolvimento , Humanos , Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/crescimento & desenvolvimento , Camundongos , Proteínas Nucleares/metabolismo , Ligação Proteica , RNA/genética , Proteínas Repressoras/metabolismo , Retroviridae/crescimento & desenvolvimento , Proteína 28 com Motivo Tripartido , Replicação Viral
15.
J Virol ; 83(6): 2429-35, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19116259

RESUMO

Mammalian genomes harbor a large number of retroviral elements acquired as germ line insertions during evolution. Although many of the endogenous retroviruses are defective, several contain one or more intact viral genes that are expressed under certain physiological or pathological conditions. This is true of the endogenous polytropic retroviruses that generate recombinant polytropic murine leukemia viruses (MuLVs). In these recombinants the env gene sequences of exogenous ecotropic MuLVs are replaced with env gene sequences from an endogenous polytropic retrovirus. Although replication-competent endogenous polytropic retroviruses have not been observed, the recombinant polytropic viruses are capable of replicating in numerous species. Recombination occurs during reverse transcription of a virion RNA heterodimer comprised of an RNA transcript from an endogenous polytropic virus and an RNA transcript from an exogenous ecotropic MuLV RNA. It is possible that homodimers corresponding to two full-length endogenous RNA genomes are also packaged. Thus, infection by an exogenous virus may result not only in recombination with endogenous sequences, but also in the mobilization of complete endogenous retrovirus genomes via pseudotyping within exogenous retroviral virions. We report that the infection of mice with an ecotropic virus results in pseudotyping of intact endogenous viruses that have not undergone recombination. The endogenous retroviruses infect and are integrated into target cell genomes and subsequently replicate and spread as pseudotyped viruses. The mobilization of endogenous retroviruses upon infection with an exogenous retrovirus may represent a major interaction of exogenous retroviruses with endogenous retroviruses and may have profound effects on the pathogenicity of retroviral infections.


Assuntos
Retrovirus Endógenos/crescimento & desenvolvimento , Vírus da Leucemia Murina/crescimento & desenvolvimento , Montagem de Vírus , Animais , Linhagem Celular , DNA Viral/química , DNA Viral/genética , Retrovirus Endógenos/fisiologia , Vírus da Leucemia Murina/fisiologia , Camundongos , Dados de Sequência Molecular , Recombinação Genética , Análise de Sequência de DNA
16.
Vaccine ; 25(52): 8660-3, 2007 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-18023943

RESUMO

EcoHIV/NL4-3 is a chimeric human immunodeficiency virus type 1 (HIV-1) that can productively infect mice. This study tests the utility of EcoHIV/NL4-3 infection to reveal protective immune responses to an HIV-1 vaccine. Immunocompetent mice were first immunized with VRC 4306 which encodes subtype B consensus sequences of gag, pol, and nef and then were infected by EcoHIV/NL4-3. Anti-Gag antibodies were sampled during immunization and infection. The extent of EcoHIV/NL4-3 infection in spleen cells and peritoneal macrophages was determined by quantitative real-time PCR (QPCR). Although antibody titres were not significantly different in control and vaccinated groups, VRC 4306 immunization induced protective responses that significantly reduced virus burden in both lymphocyte and macrophage compartments. These results indicate that EcoHIV/NL4-3 infection can be controlled by HIV-1 vaccine-induced responses, introducing a small animal model to test vaccine efficacy against HIV-1 infection.


Assuntos
Vacinas contra a AIDS/imunologia , Modelos Animais de Doenças , Infecções por HIV/prevenção & controle , HIV-1/crescimento & desenvolvimento , Vírus da Leucemia Murina/crescimento & desenvolvimento , Animais , Feminino , Anticorpos Anti-HIV/sangue , Infecções por HIV/imunologia , HIV-1/genética , Vírus da Leucemia Murina/genética , Linfócitos/virologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , RNA Viral/genética , Recombinação Genética , Baço/virologia
17.
Genome Biol ; 8(4): R48, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17407599

RESUMO

BACKGROUND: Host factors required for retroviral infection are potential targets for the modulation of diseases caused by retroviruses. During the retroviral life cycle, numerous cellular factors interact with the virus and play an essential role in infection. Cultured embryonic stem (ES) cells are susceptible to retroviral infection, therefore providing access to all of the genes required for this process to take place. In order to identify the host factors involved in retroviral infection, we designed and implemented a scheme for identifying ES cells that are resistant to retroviral infection and subsequent cloning of the mutated gene. RESULTS: A library of mutant ES cells was established by genome-wide insertional mutagenesis in Blm-deficient ES cells, and a screen was performed by superinfection of the library at high multiplicity with a recombinant retrovirus carrying a positive and negative selection cassette. Stringent negative selection was then used to exclude the infected ES cells. We successfully recovered five independent clones of ES cells that are resistant to retroviral infection. Analysis of the mutations in these clones revealed four different homozygous and one compound heterozygous mutation in the mCat-1 locus, which confirms that mCat-1 is the ecotropic murine leukemia virus receptor in ES cells. CONCLUSION: We have demonstrated the feasibility and reliability of this recessive genetic approach to identifying critical genes required for retroviral infection in ES cells; the approach provides a unique opportunity to recover other cellular factors required for retroviral infection. The resulting insertionally mutated Blm-deficient ES cell library might also provide access to essential host cell components that are required for infection and replication for other types of virus.


Assuntos
Adenosina Trifosfatases/genética , Canais de Cálcio/genética , DNA Helicases/genética , Células-Tronco Embrionárias/virologia , Vírus da Leucemia Murina/crescimento & desenvolvimento , Vírus da Leucemia Murina/genética , Mutagênese Insercional , Receptores Virais/genética , Canais de Cátion TRPV/genética , Animais , Canais de Cálcio/fisiologia , Células Cultivadas , Clonagem Molecular , Genes Recessivos , Marcadores Genéticos , Vetores Genéticos , Imunidade Inata/genética , Integrases , Camundongos , RecQ Helicases , Canais de Cátion TRPV/fisiologia
18.
PLoS Pathog ; 3(12): e200, 2007 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-18166079

RESUMO

TRIM5alpha is a restriction factor that limits infection of human cells by so-called N- but not B- or NB-tropic strains of murine leukemia virus (MLV). Here, we performed a mutation-based functional analysis of TRIM5alpha-mediated MLV restriction. Our results reveal that changes at tyrosine(336) of human TRIM5alpha, within the variable region 1 of its C-terminal PRYSPRY domain, can expand its activity to B-MLV and to the NB-tropic Moloney MLV. Conversely, we demonstrate that the escape of MLV from restriction by wild-type or mutant forms of huTRIM5alpha can be achieved through interdependent changes at positions 82, 109, 110, and 117 of the viral capsid. Together, our results support a model in which TRIM5alpha-mediated retroviral restriction results from the direct binding of the antiviral PRYSPRY domain to the viral capsid, and can be prevented by interferences exerted by critical residues on either one of these two partners.


Assuntos
Proteínas de Transporte/metabolismo , Fibroblastos/virologia , Vírus da Leucemia Murina/crescimento & desenvolvimento , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Substituição de Aminoácidos , Animais , Fatores de Restrição Antivirais , Capsídeo/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/genética , Células , Fibroblastos/citologia , Fibroblastos/imunologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/genética , HIV-1/crescimento & desenvolvimento , Humanos , Rim/citologia , Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/imunologia , Camundongos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Infecções por Retroviridae/imunologia , Proteínas com Motivo Tripartido , Infecções Tumorais por Vírus/imunologia , Ubiquitina-Proteína Ligases
19.
Virol J ; 3: 73, 2006 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-16956408

RESUMO

BACKGROUND: Retroviral Gag determines virus assembly at the plasma membrane and the formation of virus-like particles in intracellular multivesicular bodies. Thereby, retroviruses exploit by interaction with cellular partners the cellular machineries for vesicular transport in various ways. RESULTS: The retroviral Gag precursor protein drives assembly of murine leukaemia viruses (MLV) at the plasma membrane (PM) and the formation of virus like particles in multivesicular bodies (MVBs). In our study we show that caveolin-1 (Cav-1), a multifunctional membrane-associated protein, co-localizes with Gag in a punctate pattern at the PM of infected NIH 3T3 cells. We provide evidence that Cav-1 interacts with the matrix protein (MA) of the Gag precursor. This interaction is mediated by a Cav-1 binding domain (CBD) within the N-terminus of MA. Interestingly, the CBD motif identified within MA is highly conserved among most other gamma-retroviruses. Furthermore, Cav-1 is incorporated into MLV released from NIH 3T3 cells. Overexpression of a GFP fusion protein containing the putative CBD of the retroviral MA resulted in a considerable decrease in production of infectious retrovirus. Moreover, expression of a dominant-negative Cav-1 mutant affected retroviral titres significantly. CONCLUSION: This study demonstrates that Cav-1 interacts with MLV Gag, co-localizes with Gag at the PM and affects the production of infectious virus. The results strongly suggest a role for Cav-1 in the process of virus assembly.


Assuntos
Caveolina 1/metabolismo , Membrana Celular/ultraestrutura , Produtos do Gene gag/metabolismo , Vírus da Leucemia Murina/fisiologia , Animais , Vírus da Leucemia Murina/crescimento & desenvolvimento , Camundongos , Células NIH 3T3 , Replicação Viral
20.
Virology ; 354(1): 15-27, 2006 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-16887163

RESUMO

TRIM5alpha acts on several retroviruses, including human immunodeficiency virus (HIV-1), to restrict cross-species transmission. Using natural history cohorts and tissue culture systems, we examined the effect of polymorphism in human TRIM5alpha on HIV-1 infection. In African Americans, the frequencies of two non-coding SNP variant alleles in exon 1 and intron 1 of TRIM5 were elevated in HIV-1-infected persons compared with uninfected subjects. By contrast, the frequency of the variant allele encoding TRIM5alpha 136Q was relatively elevated in uninfected individuals, suggesting a possible protective effect. TRIM5alpha 136Q protein exhibited slightly better anti-HIV-1 activity in tissue culture than the TRIM5alpha R136 protein. The 43Y variant of TRIM5alpha was less efficient than the H43 variant at restricting HIV-1 and murine leukemia virus infections in cultured cells. The ancestral TRIM5 haplotype specifying no observed variant alleles appeared to be protective against infection, and the corresponding wild-type protein partially restricted HIV-1 replication in vitro. A single logistic regression model with a permutation test indicated the global corrected P value of <0.05 for both SNPs and haplotypes. Thus, polymorphism in human TRIM5 may influence susceptibility to HIV-1 infection, a possibility that merits additional evaluation in independent cohorts.


Assuntos
Proteínas de Transporte/genética , Infecções por HIV/genética , Infecções por HIV/imunologia , Polimorfismo de Nucleotídeo Único , Adolescente , Adulto , Negro ou Afro-Americano/genética , Substituição de Aminoácidos , Animais , Fatores de Restrição Antivirais , Proteínas de Transporte/fisiologia , Linhagem Celular , Criança , Pré-Escolar , Suscetibilidade a Doenças , Cães , Éxons , Frequência do Gene , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , HIV-1/crescimento & desenvolvimento , Haplótipos , Humanos , Lactente , Recém-Nascido , Íntrons , Vírus da Leucemia Murina/crescimento & desenvolvimento , Modelos Logísticos , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA