Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Gene Med ; 16(7-8): 166-78, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25044583

RESUMEN

BACKGROUND: Gene transfer technologies have the potential to fundamentally improve current therapies for arthritic conditions, although this is essentially dependent on safe and efficient vector systems. The foamy virus (FV)-based vectors have many safety features that favour their use in the treatment of arthritis. In the present study, we investigated the use of safe prototype foamy viral vectors (FVV) for indirect gene delivery to articular tissues. METHODS: We generated recombinant FVV encoding enhanced green fluorescent protein (EGFP) or human interleukin 1 receptor antagonist protein (IL1RA) cDNA under the control of the spleen focus forming virus U3 promoter and explored their transgene expression profile following ex vivo gene delivery to knee joints of Wistar and athymic nude rats. RESULTS: FVV efficiently transduced primary rat synovial fibroblasts using the EGFP and the IL1RA transgene in vitro. FVV-mediated IL1RA expression was functional in blocking IL1 effects in vitro. After the transplantation of FVV transduced synovial fibroblasts, the intra-articular transgene expression in Wistar rats was initially high and declined after approximately 3 weeks for both transgenes. By contrast, FVV-mediated expression of EGFP and IL1RA persisted for at least 12 weeks at high levels in immunocompromised nude rats. FVV-meditated gene delivery was well tolerated by all animals without extra-articular transgene expression, arguing for the safety of this approach. CONCLUSIONS: Our results indicate that FVV are capable of efficient ex vivo gene transfer to synovium and merit further investigation as a means to provide long-term intra-articular transgene expression for arthritis treatment.


Asunto(s)
Spumavirus/genética , Membrana Sinovial/citología , Animales , Células Cultivadas , Fibroblastos/metabolismo , Expresión Génica , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Humanos , Proteína Antagonista del Receptor de Interleucina 1/biosíntesis , Proteína Antagonista del Receptor de Interleucina 1/genética , Articulación de la Rodilla/citología , Articulación de la Rodilla/metabolismo , Masculino , Ratas Desnudas , Ratas Wistar , Transducción Genética
2.
ScientificWorldJournal ; 2014: 487969, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25009830

RESUMEN

Retroviral vectors are potent tools for gene delivery and various biomedical applications. To accomplish a gene transfer task successfully, retroviral vectors must effectively transduce diverse cell cultures at different phases of a cell cycle. However, very promising retroviral vectors based on the foamy viral (FV) backbone lack the capacity to efficiently transduce quiescent cells. It is hypothesized that this phenomenon might be explained as the inability of foamy viruses to form a pre-integration complex (PIC) with nuclear import activity in growth-arrested cells, which is the characteristic for lentiviruses (HIV-1). In this process, the HIV-1 central polypurine tract (cPPT) serves as a primer for plus-strand synthesis to produce a "flap" element and is believed to be crucial for the subsequent double-stranded cDNA formation of all retroviral RNA genomes. In this study, the effects of the lentiviral cPPT element on the FV transduction potential in dividing and growth-arrested (G1/S phase) adenocarcinomic human alveolar basal epithelial (A549) cells are investigated by experimental and theoretical methods. The results indicated that the HIV-1 cPPT element in a foamy viral vector background will lead to a significant reduction of the FV transduction and viral titre in growth-arrested cells due to the absence of PICs with nuclear import activity.


Asunto(s)
División Celular/fisiología , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Inhibidores de Crecimiento/administración & dosificación , VIH-1/genética , Virus Espumoso de los Simios/genética , División Celular/efectos de los fármacos , Línea Celular Tumoral , Predicción , Humanos , Lentivirus/genética
3.
J Gene Med ; 15(3-4): 155-67, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23554302

RESUMEN

BACKGROUND: Genetic treatments of chronic arthritic conditions are essentially dependent on safe and efficient vector systems. To combine features of the efficient transduction of adenovirus vectors with the advantage of stable integration into the host cell genome of apathogenic prototype foamy virus vectors, hybrid vectors (FAD) have been established. In the present study, we have generated and investigated the use of safe FAD vectors for direct gene delivery to joints. METHODS: We generated recombinant FAD encoding enhanced green fluorescent protein (EGFP) or human interleukin 1 receptor antagonist protein (IL1RA) cDNA, and explored their transgene expression profile, as well as the bioactivity of the IL1RA transgene in vitro. The feasibility of IL1RA gene delivery to articular tissues was investigated in a pilot study employing direct FAD injections to the knee joints of Wistar rats. RESULTS: FAD vectors efficiently transduced human or rat fibroblasts with EGFP or IL1RA transgene in vitro. Levels of IL1RA transgene expression were high, stable and functional in vitro. Transduced synovial fibroblasts and high levels of IL1RA protein (10-35 ng/ml) could be detected in vivo in the synovium of Wistar rats 3-5 days after injection of FAD vectors to the knee joints. CONCLUSIONS: Our results indicate that FAD vectors are capable of efficient in vivo gene transfer to synovium and merit further investigation as a means of providing efficient and long-term intra-articular transgene expression for treatment of the arthritides.


Asunto(s)
Adenoviridae/genética , Artritis/terapia , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Spumavirus/genética , Membrana Sinovial/metabolismo , Animales , Western Blotting , Línea Celular , Cartilla de ADN/genética , Electroforesis en Gel de Poliacrilamida , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Proteína Antagonista del Receptor de Interleucina 1/genética , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Proyectos Piloto , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transgenes/genética
4.
J Virol ; 86(18): 10028-35, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22787203

RESUMEN

The cellular receptor of foamy viruses (FVs) is unknown. The broad spectrum of permissive cells suggests that the cellular receptor is a molecular structure with almost ubiquitous prevalence. Here, we investigated the ability of heparan sulfate (HS), a glycosaminoglycan (GAG) present on the extracellular matrix of many cells, to bind FV particles and to permit prototype FV (PFV) and feline FV (FFV) entry. Permissivity of different cell lines for FV entry correlated with the amount of heparan sulfate present on the cell surface. The resulting 50% cell culture infectious doses (CCID(50)s) were distributed over a range of 4 logs, which means that the most susceptible cell line tested (HT1080) was more than 10,000 times more susceptible for PFV infection than the least susceptible cell line (CRL-2242). HS surface expression varied over a range of 2 logs. HS expression and FV susceptibility were positively correlated (P < 0.001). Enzymatic digestion of heparan sulfate on HT1080 cells diminished permissivity for PFV entry by a factor of at least 500. Using fast protein liquid chromatography (FPLC), we demonstrated binding of FV vector particles to a gel filtration column packed with heparin, a molecule structurally related to heparan sulfate, allowing for the purification of infectious particles. Both PFV and FFV infection were inhibited by soluble heparin. Our results show that FVs bind to HS and that this interaction is a pivotal step for viral entry, suggesting that HS is a cellular attachment factor for FVs.


Asunto(s)
Heparitina Sulfato/fisiología , Spumavirus/fisiología , Acoplamiento Viral , Animales , Gatos , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , Membrana Celular/virología , Cricetinae , Progresión de la Enfermedad , Heparina/metabolismo , Heparina/farmacología , Heparitina Sulfato/deficiencia , Humanos , Ratones , Receptores Virales/efectos de los fármacos , Receptores Virales/fisiología , Infecciones por Retroviridae/prevención & control , Spumavirus/patogenicidad , Acoplamiento Viral/efectos de los fármacos , Internalización del Virus/efectos de los fármacos
5.
Retrovirology ; 9: 41, 2012 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-22574974

RESUMEN

BACKGROUND: Recently, contradictory results on foamy virus protease activity were published. While our own results indicated that protease activity is regulated by the viral RNA, others suggested that the integrase is involved in the regulation of the protease. RESULTS: To solve this discrepancy we performed additional experiments showing that the protease-reverse transcriptase (PR-RT) exhibits protease activity in vitro and in vivo, which is independent of the integrase domain. In contrast, Pol incorporation, and therefore PR activity in the viral context, is dependent on the integrase domain. To further analyse the regulation of the protease, we incorporated Pol in viruses by expressing a GagPol fusion protein, which supported near wild-type like infectivity. A GagPR-RT fusion, lacking the integrase domain, also resulted in wild-type like Gag processing, indicating that the integrase is dispensable for viral Gag maturation. Furthermore, we demonstrate with a trans-complementation assays that the PR in the context of the PR-RT protein supports in trans both, viral maturation and infectivity. CONCLUSION: We provide evidence that the FV integrase is required for Pol encapsidation and that the FV PR activity is integrase independent. We show that an active PR can be encapsidated in trans as a GagPR-RT fusion protein.


Asunto(s)
Ácido Aspártico Endopeptidasas/química , Proteínas de Fusión gag-pol/química , Regulación Enzimológica de la Expresión Génica , Regulación Viral de la Expresión Génica , Integrasas/química , ARN Viral/química , Virus Espumoso de los Simios/enzimología , Activación Enzimática , Proteínas de Fusión gag-pol/genética , Prueba de Complementación Genética , Células HEK293 , Humanos , Plásmidos/química , Plásmidos/genética , Estructura Terciaria de Proteína , Proteolisis , ARN Viral/genética , ADN Polimerasa Dirigida por ARN/química , Virus Espumoso de los Simios/química , Virus Espumoso de los Simios/genética , Transfección , Proteínas Virales/química , Proteínas Virales/genética
6.
J Virol ; 85(5): 2333-41, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21159877

RESUMEN

Most retroviruses express all of their genes from a single primary transcript. In order to allow expression of more than one gene from this RNA, differential splicing is extensively used. Cellular quality control mechanisms retain and degrade unspliced or partially spliced RNAs in the nucleus. Two pathways have been described that explain how retroviruses circumvent this nuclear export inhibition. One involves a constitutive transport element in the viral RNA that interacts with the cellular mRNA transporter proteins NXF1 and NXT1 to facilitate nuclear export. The other pathway relies on the recognition of a viral RNA element by a virus-encoded protein that interacts with the karyopherin CRM1. In this report, we analyze the protein factors required for the nuclear export of unspliced foamy virus (FV) mRNA. We show that this export is CRM1 dependent. In contrast to other complex retroviruses, FVs do not encode an export-mediating protein. Cross-linking experiments indicated that the cellular protein HuR binds to the FV RNA. Inhibition studies showed that both ANP32A and ANP32B, which are known to bridge HuR and CRM1, are essential for FV RNA export. By using this export pathway, FVs solve a central problem of viral replication.


Asunto(s)
Núcleo Celular/metabolismo , ARN Viral/metabolismo , Infecciones por Retroviridae/virología , Spumavirus/metabolismo , Transporte Activo de Núcleo Celular , Núcleo Celular/genética , Células HEK293 , Humanos , Carioferinas/metabolismo , Transporte de ARN , ARN Viral/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Retroviridae/genética , Retroviridae/metabolismo , Infecciones por Retroviridae/metabolismo , Spumavirus/genética , Proteína Exportina 1
7.
J Virol ; 85(9): 4462-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21325405

RESUMEN

Foamy viruses (FVs) synthesize the Pol precursor protein from a specific transcript. Thus, in contrast to what was found for orthoretroviruses, e.g., human immunodeficiency virus, no Gag-Pol precursor protein is synthesized. Foamy viral Pol consists of a protease (PR) domain, a reverse transcriptase domain, and an integrase domain and is processed into a mature protease-reverse transcriptase (PR-RT) fusion protein and the integrase. Protease activity has to be strictly regulated in order to avoid premature Gag and Pol processing before virus assembly. We have demonstrated recently that FV protease is an inactive monomer with a very weak dimerization tendency and postulated protease activation through dimerization. Here, we identify a specific protease-activating RNA motif (PARM) located in the pol region of viral RNA which stimulates PR activity in vitro and in vivo, revealing a novel and unique mechanism of retroviral protease activation. This mechanism is strikingly different to that of orthoretroviruses, where the protease can be activated even in the absence of viral RNA during the assembly of virus-like particles. Although it has been shown that the integrase domain is important for Pol uptake, activation of the foamy virus protease is integrase independent. We show that at least two foamy virus PR-RT molecules bind to the PARM and only RNAs containing the PARM result in significant activation of the protease. DNA harboring the PARM is not capable of protease activation. Structure determination of the PARM by selective 2' hydroxyl acylation analyzed by primer extension (SHAPE) revealed a distinct RNA folding, important for protease activation and thus virus maturation.


Asunto(s)
Ácido Aspártico Endopeptidasas/metabolismo , Regulación Viral de la Expresión Génica , ARN Viral/metabolismo , Virus Espumoso de los Simios/fisiología , Humanos , Conformación de Ácido Nucleico , ARN Viral/química
8.
J Virol ; 85(8): 3986-95, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21289113

RESUMEN

Foamy virus (FV) capsid proteins have few lysines. Basic residues are almost exclusively represented by arginines indicating positive selective pressure. To analyze the possible functions of this peculiarity, we mutated an infectious molecular clone of the prototypic FV (PFV) to harbor lysines in the Gag protein at arginine-specifying positions and analyzed various aspects of the FV replication cycle. The majority of mutants replicated equally as well in permanent cell cultures as the original wild-type (wt) virus and were genetically stable in gag upon 10 cell-free passages. With respect to the features of late reverse transcription, nucleic acid content, and infectiousness of the virion DNA genome, the majority of mutants behaved like the wt. Several mutants of PFV were ubiquitinated in Gag but unable to generate virus-like particles (VLPs) or to undergo pseudotyping by a heterologous envelope. Using primary cells, however, a replicative disadvantage of the majority of mutants was disclosed. This disadvantage was enhanced upon interferon (IFN) treatment. We found no evidence that the lysine-bearing gag mutants showed more restriction than the wt virus by tetherin (CD317) or Trim5α. A single lysine in PFV Gag was found to be nonessential for transient replication in permanent cell culture if replaced by an arginine residue. Upon replication in primary cells, even without IFN treatment, this mutant was severely impaired, indicating the importance of specifying at least this lysine residue in PFV Gag. The paucity of lysines in FV Gag proteins may be a consequence of preventing proteasomal Gag degradation.


Asunto(s)
Aminoácidos Básicos/metabolismo , Productos del Gen gag/química , Spumavirus/química , Spumavirus/fisiología , Aminoácidos Básicos/genética , Productos del Gen gag/genética , Mutagénesis Sitio-Dirigida , Spumavirus/genética , Virulencia , Replicación Viral
9.
BMC Infect Dis ; 12: 14, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22264238

RESUMEN

BACKGROUND: HIV-associated general immune activation is a strong predictor for HIV disease progression, suggesting that chronic immune activation may drive HIV pathogenesis. Consequently, immunomodulating agents may decelerate HIV disease progression. METHODS: In an observational study, we determined immune activation in HIV patients receiving low-dose (5 mg/day) prednisolone with or without highly-active antiretroviral therapy (HAART) compared to patients without prednisolone treatment. Lymphocyte activation was determined by flow cytometry detecting expression of CD38 on CD8(+) T cells. The monocyte activation markers sCD14 and LPS binding protein (LBP) as well as inflammation markers soluble urokinase plasminogen activated receptor (suPAR) and sCD40L were determined from plasma by ELISA. RESULTS: CD38-expression on CD8+ T lymphocytes was significantly lower in prednisolone-treated patients compared to untreated patients (median 55.40% [percentile range 48.76-67.70] versus 73.34% [65.21-78.92], p = 0.0011, Mann-Whitney test). Similarly, we detected lower levels of sCD14 (3.6 µg/ml [2.78-5.12] vs. 6.11 µg/ml [4.58-7.70]; p = 0.0048), LBP (2.18 ng/ml [1.59-2.87] vs. 3.45 ng/ml [1.84-5.03]; p = 0.0386), suPAR antigen (2.17 µg/ml [1.65-2.81] vs. 2.56 µg/ml [2.24-4.26]; p = 0.0351) and a trend towards lower levels of sCD40L (2.70 pg/ml [1.90-4.00] vs. 3.60 pg/ml [2.95-5.30]; p = 0.0782). Viral load in both groups was similar (0.8 × 105 ng/ml [0.2-42.4 × 105] vs. 1.1 × 105 [0.5-12.2 × 105]; p = 0.3806). No effects attributable to prednisolone were observed when patients receiving HAART in combination with prednisolone were compared to patients who received HAART alone. CONCLUSIONS: Patients treated with low-dose prednisolone display significantly lower general immune activation than untreated patients. Further longitudinal studies are required to assess whether treatment with low-dose prednisolone translates into differences in HIV disease progression.


Asunto(s)
Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , Factores Inmunológicos/administración & dosificación , Activación de Linfocitos/efectos de los fármacos , Prednisolona/administración & dosificación , ADP-Ribosil Ciclasa 1/análisis , Proteínas de Fase Aguda/análisis , Adulto , Fármacos Anti-VIH/administración & dosificación , Ligando de CD40/sangre , Linfocitos T CD8-positivos/química , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Proteínas Portadoras/análisis , Femenino , Citometría de Flujo , Humanos , Inmunofenotipificación , Receptores de Lipopolisacáridos/análisis , Masculino , Glicoproteínas de Membrana/análisis , Persona de Mediana Edad , Monocitos/química , Monocitos/inmunología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/sangre
10.
Bioorg Med Chem ; 20(17): 5277-89, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22858300

RESUMEN

Malaria and HIV are among the most important global health problems of our time and together are responsible for approximately 3 million deaths annually. These two diseases overlap in many regions of the world including sub-Saharan Africa, Southeast Asia and South America, leading to a higher risk of co-infection. In this study, we generated and characterized hybrid molecules to target Plasmodium falciparum and HIV simultaneously for a potential HIV/malaria combination therapy. Hybrid molecules were synthesized by the covalent fusion of azidothymidine (AZT) with dihydroartemisinin (DHA), a tetraoxane or a 4-aminoquinoline derivative; and the small library was tested for antiviral and antimalarial activity. Our data suggests that compound 7 is the most potent molecule in vitro, with antiplasmodial activity comparable to that of DHA (IC(50)=26 nM, SI>3000), a moderate activity against HIV (IC(50)=2.9 µM; SI>35) and not toxic to HeLa cells at concentrations used in the assay (CC(50)>100 µM). Pharmacokinetics studies further revealed that compound 7 is metabolically unstable and is cleaved via O-dealkylation. These studies account for the lack of in vivo efficacy of compound 7 against the CQ-sensitive Plasmodium berghei N strain in mice, when administered orally at 20mg/kg.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/tratamiento farmacológico , Fármacos Anti-VIH/farmacología , Antimaláricos/farmacología , Infecciones por VIH/tratamiento farmacológico , Malaria/tratamiento farmacológico , Plasmodium falciparum/efectos de los fármacos , Animales , Fármacos Anti-VIH/administración & dosificación , Fármacos Anti-VIH/síntesis química , Antimaláricos/administración & dosificación , Antimaláricos/síntesis química , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , VIH-1/efectos de los fármacos , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Pruebas de Sensibilidad Parasitaria , Relación Estructura-Actividad
11.
J Gen Virol ; 92(Pt 3): 582-6, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21106805

RESUMEN

To shed light on primate foamy virus (FV) evolution, we determined the complete nucleotide sequence of the gorilla simian foamy virus (SFVgor). Starting from a conserved region in the integrase (IN) domain of the pol gene we cloned the viral genome to the 5' and 3' LTR into plasmid vectors and elucidated its nucleotide sequence. The sequences of both LTRs were determined by nucleotide sequencing of separate PCR products from the primer-binding site or the bel region and LTRs. All protein motifs conserved among the primate FV were identified in SFVgor. Using phylogenetic analysis of the Gag, Pol and Env amino acid sequences, we demonstrate that SFVgor consistently clusters in accordance with a scenario of virus-host co-divergence.


Asunto(s)
Secuencia de Bases , Genoma Viral , Gorilla gorilla/virología , Virus Espumoso de los Simios/genética , Secuencias de Aminoácidos , Animales , Clonación Molecular , Análisis por Conglomerados , Secuencia Conservada , Productos del Gen env/genética , Productos del Gen gag/genética , Productos del Gen pol/genética , Vectores Genéticos , Datos de Secuencia Molecular , Filogenia , Plásmidos , ARN Viral/genética , Análisis de Secuencia de ADN , Homología de Secuencia , Virus Espumoso de los Simios/aislamiento & purificación
12.
Exp Cell Res ; 316(11): 1885-95, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20171964

RESUMEN

Many regenerative cells are label-retaining cells (LRCs) due to their ability to keep a DNA label over a prolonged time. Until recently, isolation of vital LRCs was hampered due to the necessary use of fixation methods. To circumvent this, we generated a lentiviral-(HIV-1) based vector expressing a Tet-Off controlled histone 2B-GFP (Tet-Off-H2B-GFP) reporter gene for the detection and isolation of viable LRCs. In initial experiments, the vector was successfully used to infect 2- and 3-dimensional tissue culture models. Infected cultures from skin and pancreatic cells showed a very tight regulation of H2B-GFP, were sensitive to minimal amounts of doxycycline (Dox) and had a stable transgenic expression over the time of this study. Our lentiviral vector represents a reliable and easy to handle system for the successful infection, detection and isolation of LRCs from various tissues in vitro, in vivo and ex vivo.


Asunto(s)
Genes Reporteros , Vectores Genéticos , Histonas/genética , Lentivirus/genética , Animales , Ciclo Celular , Línea Celular , Doxiciclina/farmacología , Expresión Génica/efectos de los fármacos , Ingeniería Genética , Proteínas Fluorescentes Verdes/genética , Humanos , Ratones , Proteínas Recombinantes de Fusión/genética , Transfección
13.
Retrovirology ; 7: 5, 2010 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-20113504

RESUMEN

BACKGROUND: The foamy virus Pol protein is translated independently from Gag using a separate mRNA. Thus, in contrast to orthoretroviruses no Gag-Pol precursor protein is synthesized. Only the integrase domain is cleaved off from Pol resulting in a mature reverse transcriptase harboring the protease domain at the N-terminus (PR-RT). Although the homology between the PR-RTs from simian foamy virus from macaques (SFVmac) and the prototype foamy virus (PFV), probably originating from chimpanzee, exceeds 90%, several differences in the biophysical and biochemical properties of the two enzymes have been reported (i.e. SFVmac develops resistance to the nucleoside inhibitor azidothymidine (AZT) whereas PFV remains AZT sensitive even if the resistance mutations from SFVmac PR-RT are introduced into the PFV PR-RT gene). Moreover, contradictory data on the monomer/dimer status of the foamy virus protease have been published. RESULTS: We set out to purify and directly compare the monomer/dimer status and the enzymatic behavior of the two wild type PR-RT enzymes from SFVmac and PFV in order to get a better understanding of the protein and enzyme functions. We determined kinetic parameters for the two enzymes, and we show that PFV PR-RT is also a monomeric protein. CONCLUSIONS: Our data show that the PR-RTs from SFV and PFV are monomeric proteins with similar biochemical and biophysical properties that are in some aspects comparable with MLV RT, but differ from those of HIV-1 RT. These differences might be due to the different conditions the viruses are confronted with in dividing and non-dividing cells.


Asunto(s)
ADN Polimerasa Dirigida por ARN/química , Spumavirus/enzimología , Proteínas Virales/química , Proteínas Virales/metabolismo , Cromatografía en Gel , Dicroismo Circular , Cinética , Peso Molecular , Pliegue de Proteína , Multimerización de Proteína , ADN Polimerasa Dirigida por ARN/metabolismo , Proteínas Virales/aislamiento & purificación
14.
Antimicrob Agents Chemother ; 54(1): 78-87, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19841153

RESUMEN

Inhibition of the interaction of the human cytidine-deaminase APOBEC3G (A3G) with the human immunodeficiency virus (HIV) type 1-specific viral infectivity factor (Vif) represents a novel therapeutic approach in which a cellular factor with potent antiviral activity (A3G) plays a key role. In HIV-infected cells, the interaction of Vif with A3G leads to the subsequent degradation of A3G by the 26S proteasome via the ubiquitin pathway and to the loss of antiviral activity. To establish a stable and convenient cellular testing platform for the high-throughput screening of potential antiviral compound libraries, we engineered a double transgenic cell line constitutively expressing an enhanced yellow fluorescent protein expressor (EYFP-A3G) fusion as well as a Tet-Off controllable Vif protein. With this cell line, we were able to measure precisely the Vif-induced degradation of A3G in the presence of potential antiviral compounds in an easy-to-handle, robust, and practical high-throughput multiwell plate format with an excellent screening window coefficient (Z factor) of 0.67.


Asunto(s)
Fármacos Anti-VIH/farmacología , Antivirales/farmacología , Citidina Desaminasa/genética , Evaluación Preclínica de Medicamentos/métodos , Antibacterianos/farmacología , Fármacos Anti-VIH/síntesis química , Fármacos Anti-VIH/economía , Antivirales/síntesis química , Antivirales/economía , Secuencia de Bases , Western Blotting , Línea Celular , Análisis Costo-Beneficio , Medios de Cultivo , Citidina Desaminasa/antagonistas & inhibidores , Citidina Desaminasa/biosíntesis , ADN Viral/genética , Doxiciclina/farmacología , Evaluación Preclínica de Medicamentos/economía , Electroforesis en Gel de Poliacrilamida , Inducción Enzimática/efectos de los fármacos , Citometría de Flujo , Colorantes Fluorescentes , Regulación Viral de la Expresión Génica/efectos de los fármacos , Vectores Genéticos , Humanos , Lentivirus/efectos de los fármacos , Datos de Secuencia Molecular , Plásmidos/genética , Reproducibilidad de los Resultados , Transfección
15.
Med Microbiol Immunol ; 199(3): 197-207, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20445989

RESUMEN

One of the most fascinating areas in retrovirology is the study of foamy viruses (FVs), because these viruses appear to do everything that is common to all other retroviruses differently. FVs have found a completely new way to propagate their genome. And they do this extremely successfully because most of wild non-human primates, felines, bovines, equines, and small ruminants are likely to be non-pathogenically infected. The success of FVs can also be viewed from a different angle, since they replicate very conservatively and do not need to shape their genotypic and phenotypic makeup every now and then. The elucidation of the underlying basic mechanisms of the FV replication strategy is the topic of this review.


Asunto(s)
Biología Molecular , Virus Espumoso de los Simios/fisiología , Replicación Viral , Animales , Humanos , Modelos Biológicos , Virus Espumoso de los Simios/genética , Virus Espumoso de los Simios/crecimiento & desarrollo , Virus Espumoso de los Simios/patogenicidad
16.
Nucleic Acids Res ; 36(3): 1009-16, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18096624

RESUMEN

Azidothymidine (AZT, zidovudine) is one of the few nucleoside inhibitors known to inhibit foamy virus replication. We have shown previously that up to four mutations in the reverse transcriptase gene of simian foamy virus from macaque (SFVmac) are necessary to confer high resistance against AZT. To characterize the mechanism of AZT resistance we expressed two recombinant reverse transcriptases of highly AZT-resistant SFVmac in Escherichia coli harboring three (K211I, S345T, E350K) or four mutations (K211I, I224T, S345T, E350K) in the reverse transcriptase gene. Our analyses show that the polymerization activity of these mutants is impaired. In contrast to the AZT-resistant reverse transcriptase of HIV-1, the AZT resistant enzymes of SFVmac reveal differences in their kinetic properties. The SFVmac enzymes exhibit lower specific activities on poly(rA)/oligo(dT) and higher K(M)-values for polymerization but no change in K(D)-values for DNA/DNA or RNA/DNA substrates. The AZT resistance of the mutant enzymes is based on the excision of the incorporated inhibitor in the presence of ATP. The additional amino acid change of the quadruple mutant appears to be important for regaining polymerization efficiency.


Asunto(s)
Adenosina Trifosfato/metabolismo , Didesoxinucleótidos/metabolismo , ADN Polimerasa Dirigida por ARN/genética , ADN Polimerasa Dirigida por ARN/metabolismo , Inhibidores de la Transcriptasa Inversa/farmacología , Spumavirus/enzimología , Nucleótidos de Timina/metabolismo , Zidovudina/análogos & derivados , Zidovudina/farmacología , Secuencia de Aminoácidos , ADN/biosíntesis , Farmacorresistencia Viral/genética , Escherichia coli/genética , Cinética , Datos de Secuencia Molecular , Mutación , ARN , ADN Polimerasa Dirigida por ARN/química , Alineación de Secuencia , Spumavirus/efectos de los fármacos , Zidovudina/metabolismo
17.
Retrovirology ; 6: 32, 2009 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-19348676

RESUMEN

BACKGROUND: Foamy viruses (FVs) are the most genetically stable viruses of the retrovirus family. This is in contrast to the in vitro error rate found for recombinant FV reverse transcriptase (RT). To investigate the accuracy of FV genome copying in vivo we analyzed the occurrence of mutations in HEK 293T cell culture after a single round of reverse transcription using a replication-deficient vector system. Furthermore, the frequency of FV recombination by template switching (TS) and the cross-packaging ability of different FV strains were analyzed. RESULTS: We initially sequenced 90,000 nucleotides and detected 39 mutations, corresponding to an in vivo error rate of approximately 4 x 10-4 per site per replication cycle. Surprisingly, all mutations were transitions from G to A, suggesting that APOBEC3 activity is the driving force for the majority of mutations detected in our experimental system. In line with this, we detected a late but significant APOBEC3G and 3F mRNA by quantitative PCR in the cells. We then analyzed 170,000 additional nucleotides from experiments in which we co-transfected the APOBEC3-interfering foamy viral bet gene and observed a significant 50% drop in G to A mutations, indicating that APOBEC activity indeed contributes substantially to the foamy viral replication error rate in vivo. However, even in the presence of Bet, 35 out of 37 substitutions were G to A, suggesting that residual APOBEC activity accounted for most of the observed mutations. If we subtract these APOBEC-like mutations from the total number of mutations, we calculate a maximal intrinsic in vivo error rate of 1.1 x 10-5 per site per replication. In addition to the point mutations, we detected one 49 bp deletion within the analyzed 260000 nucleotides.Analysis of the recombination frequency of FV vector genomes revealed a 27% probability for a template switching (TS) event within a 1 kilobase (kb) region. This corresponds to a 98% probability that FVs undergo at least one additional TS event per replication cycle. We also show that a given FV particle is able to cross-transfer a heterologous FV genome, although at reduced efficiency than the homologous vector. CONCLUSION: Our results indicate that the copying of the FV genome is more accurate than previously thought. On the other hand recombination among FV genomes appears to be a frequent event.


Asunto(s)
Genoma Viral/genética , Spumavirus/genética , Desaminasas APOBEC , Línea Celular , Citidina Desaminasa , Citosina Desaminasa/metabolismo , Humanos , Mutación , Proteínas de los Retroviridae/metabolismo , Moldes Genéticos , Replicación Viral
18.
Mol Ther Nucleic Acids ; 12: 626-634, 2018 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-30081233

RESUMEN

Viral vectors are rapidly being developed for a range of applications in research and gene therapy. Prototype foamy virus (PFV) vectors have been described for gene therapy, although their use has mainly been restricted to ex vivo stem cell modification. Here we report direct in vivo transgene delivery with PFV vectors carrying reporter gene constructs. In our investigations, systemic PFV vector delivery to neonatal mice gave transgene expression in the heart, xiphisternum, liver, pancreas, and gut, whereas intracranial administration produced brain expression until animals were euthanized 49 days post-transduction. Immunostaining and confocal microscopy analysis of injected brains showed that transgene expression was highly localized to hippocampal architecture despite vector delivery being administered to the lateral ventricle. This was compared with intracranial biodistribution of lentiviral vectors and adeno-associated virus vectors, which gave a broad, non-specific spread through the neonatal mouse brain without regional localization, even when administered at lower copy numbers. Our work demonstrates that PFV can be used for neonatal gene delivery with an intracranial expression profile that localizes to hippocampal neurons, potentially because of the mitotic status of the targeted cells, which could be of use for research applications and gene therapy of neurological disorders.

19.
Curr Gene Ther ; 7(4): 261-71, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17969559

RESUMEN

The first vectors derived from foamy viruses were established over ten years ago. Until now only used and further developed by a handful of investigators these vectors have been shown to be promising tools for the gene transfer into haematopoietic stem cells. Several inherent features of foamy virus-derived vectors, such as the high efficiency in targeting CD34-positive stem cells, a favourable integration profile, and the apathogenic nature of the parental virus, indicate that they are superior to gammaretroviral and lentiviral vectors. The effectiveness in different preclinical animal models suggests the exploration of foamy virus vectors in human trials.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos , Spumavirus/genética , Animales , Gammaretrovirus/genética , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/virología , Humanos , Lentivirus/genética , Modelos Animales , Virus Espumoso de los Simios/genética , Virus Espumoso de los Simios/fisiología , Spumavirus/fisiología , Ensamble de Virus , Replicación Viral
20.
Ann N Y Acad Sci ; 1091: 540-7, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17341643

RESUMEN

CpG oligodeoxynucleotides (CpG ODNs) bind to toll-like receptor-9 (TLR-9) and activate immune cells with antigen-presenting activity, including B cells and dendritic cells. Here we show that treatment of the latently human immunodeficiency virus (HIV)-infected T cell line ACH-2 with the CpG ODNs 2006 or 2040 triggers activation of viral gene expression, demonstrating that CpG-signaling activity can also be found in T cells. The CpG ODNs g12AAC and g12GTC had no effect on virus reactivation. In contrast to the stimulating effects on viral gene expression in latently infected cells, CpG ODNs potently suppressed HIV replication in productively infected MT4 T cells or PBLs. Inhibition of virus replication was not related to the CpG motif but similarly occurred with non-CpG phosphorothioate (PTO)-ODNs. Thus, virus inhibition was likely caused by the PTO backbone of the CpG ODNs, probably by interfering with events prior to integration of the viral cDNA into the host genome. The ability of CpG PTO-ODNs to trigger reactivation of latent HIV in combination with their antiviral activity on productive infection makes this substance class an interesting candidate for further test to asses their potential as supplements in HIV therapy.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Fármacos Anti-VIH/farmacología , Islas de CpG/fisiología , VIH/fisiología , Oligodesoxirribonucleótidos/farmacología , Provirus/fisiología , Linfocitos T/virología , Activación Viral/fisiología , Línea Celular , VIH/efectos de los fármacos , VIH/genética , Humanos , Provirus/efectos de los fármacos , Provirus/genética , Linfocitos T/efectos de los fármacos , Latencia del Virus/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA