Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
ChemMedChem ; 9(9): 2021-5, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24954543

RESUMEN

RNA-guided machineries perfectly satisfy the demand for rationally programmable tools that manipulate gene function inside the cell. Over the last ten years, various natural machineries have been harnessed, with RNA interference being among the most prominent examples. It is now time to tackle the engineering of novel RNA-guided tools not provided by nature. In this respect, we highlight RNA-guided site-directed RNA editing as a new concept for the manipulation of RNA and protein function. In contrast to currently available techniques, RNA editing allows for the introduction of selected point mutations into the transcriptome without the need for genomic manipulation. In particular, the approach described using chemically stabilized, antagomir-like guideRNAs may offer advantages over others, such as specificity and circumvention of immunogenicity. These new tools have significant potential for the advancement of both basic science and medicinal application, especially in the treatment of genetic diseases.


Asunto(s)
Nucleósido Desaminasas/química , Proteínas/efectos de los fármacos , Edición de ARN/efectos de los fármacos , ARN/efectos de los fármacos , Animales , Humanos , Mutagénesis Sitio-Dirigida , Nucleósido Desaminasas/farmacología , Ingeniería de Proteínas , Proteínas/fisiología , ARN/fisiología
2.
Proc Natl Acad Sci U S A ; 104(8): 2915-20, 2007 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-17299050

RESUMEN

Human T cell leukemia virus type 1 (HTLV-1) has evolved a remarkable strategy to thwart the antiviral effects of the cellular cytidine deaminase APOBEC3G (hA3G). HTLV-1 infects T lymphocytes in vivo, where, like HIV-1, it is likely to encounter hA3G. HIV-1 counteracts the innate antiviral activity of hA3G by producing an accessory protein, Vif, which hastens the degradation of hA3G. In contrast, HTLV-1 does not encode a Vif homologue; instead, HTLV-1 has evolved a cis-acting mechanism to prevent hA3G restriction. We demonstrate here that a peptide motif in the C terminus of the HTLV-1 nucleocapsid (NC) domain inhibits hA3G packaging into nascent virions. Mutation of amino acids within this region resulted in increased levels of hA3G incorporation into virions and increased susceptibility to hA3G restriction. Elements within the C-terminal extension of the NC domain are highly conserved among the primate T cell leukemia viruses, but this extension is absent in all other retroviral NC proteins.


Asunto(s)
Virus Linfotrópico T Tipo 1 Humano/efectos de los fármacos , Virus Linfotrópico T Tipo 1 Humano/fisiología , Nucleocápside/química , Nucleocápside/metabolismo , Nucleósido Desaminasas/farmacología , Proteínas Represoras/farmacología , Desaminasa APOBEC-3G , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Secuencia Conservada , Citidina Desaminasa , Productos del Gen gag/química , Productos del Gen gag/metabolismo , Productos del Gen vif/química , Productos del Gen vif/metabolismo , VIH-1 , Células HeLa , Humanos , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Nucleocápside/efectos de los fármacos , Péptidos/química , Estructura Terciaria de Proteína/efectos de los fármacos , Ensamble de Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Productos del Gen vif del Virus de la Inmunodeficiencia Humana
3.
J Immunol ; 178(3): 1671-9, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17237417

RESUMEN

Apolipoprotein B mRNA-editing, enzyme-catalytic, polypeptide-like-3G (A3G) is an intracellular innate antiviral factor that deaminates retroviral cytidine to uridine. In an attempt to harness the anti-HIV effect of A3G, we searched for an agent that would up-regulate A3G and identify the receptors involved. Stimulation of cell surface CCR5 with CCL3 and CD40 with CD40L or both molecules with microbial 70-kDa heat shock protein (HSP)70 up-regulated A3G mRNA and protein expression in human CD4(+) T cells and monocyte-derived dendritic cells (DC), demonstrated by real-time PCR and Western blots, respectively. The specificity of CCR5 and CD40 stimulation was established by inhibition with TAK 779 and mAb to CD40, as well as using human embryonic kidney 293 cells transfected with CCR5 and CD40, respectively. A dose-dependent increase of A3G in CCL3- or HSP70-stimulated CD4(+) T cells was associated with inhibition in HIV-1 infectivity. To differentiate between the inhibitory effect of HSP70-induced CCR5 binding and that of A3G, GFP-labeled pseudovirions were used to infect human embryonic kidney 293 cells, which showed inhibition of pseudovirion uptake, consistent with A3G being responsible for the inhibitory effect. Ligation of cell surface CCR5 receptors by CCL3 or CD40 by CD40L activated the ERK1/2 and p38 MAPK signaling pathways that induced A3G mRNA expression and production of the A3G protein. These in vitro results were corroborated by in vivo studies in rhesus macaques in which A3G was significantly up-regulated following immunization with SIVgp120 and p27 linked to HSP70. This novel preventive approach may in addition to adaptive immunity use the intracellular innate antiviral effect of A3G.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Antígenos CD40/metabolismo , Células Dendríticas/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Nucleósido Desaminasas/genética , Receptores CCR5/metabolismo , Proteínas Represoras/genética , Desaminasa APOBEC-3G , Animales , Fármacos Anti-VIH , Células Cultivadas , Citidina Desaminasa , Infecciones por VIH/tratamiento farmacológico , Humanos , Ligandos , Macaca mulatta , Nucleósido Desaminasas/farmacología , ARN Mensajero/farmacología , Proteínas Represoras/farmacología , Transducción de Señal , Regulación hacia Arriba/genética
4.
J Gen Virol ; 88(Pt 2): 432-440, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17251560

RESUMEN

G to A hypermutation of Hepatitis B virus (HBV) and retroviruses appears as a result of deamination activities of host APOBEC proteins and is thought to play a role in innate antiviral immunity. Alpha and gamma interferons (IFN-alpha and -gamma) have been reported to upregulate the transcription of APOBEC3G, which is known to reduce the replication of HBV. We investigated the number of hypermutated genomes under various conditions by developing a quantitative measurement. The level of hypermutated HBV in a HepG2 cell line, which is semi-permissive for retrovirus, was 2.3 in 10(4) HBV genomes, but only 0.5 in 10(4) in permissive Huh7 cells. The level of APOBEC3G mRNA was about ten times greater in HepG2 cells than in Huh7 cells. Treatment of HepG2 cells with either IFN-alpha or -gamma increased the transcription of APOBEC3G and hypermutation of HBV. These mRNAs and hypermutation of HBV genomes were induced more prominently by IFN-gamma than by IFN-alpha. Both IFNs decreased the number of replicative intermediate of HBV. Overexpression of APOBEC3G reduced the number of replicative intermediate of HBV and increased hypermutated genomes 334 times, reaching 968 in 10(4) genomes. Deamination-inactive APOBEC3G did not induce hypermutation, but reduced the virus equally. Our results suggest that APOBEC3G, upregulated by IFNs, has a dual effect on HBV: induction of hypermutation and reduction of virus synthesis. The effect of hypermutation on infectivity should be investigated further.


Asunto(s)
Virus de la Hepatitis B/efectos de los fármacos , Nucleósido Desaminasas/farmacología , Proteínas Represoras/farmacología , Desaminasa APOBEC-3G , Línea Celular , Citidina Desaminasa , ADN Viral/química , ADN Viral/genética , Genoma Viral , Virus de la Hepatitis B/genética , Interferón-alfa/farmacología , Interferón gamma/farmacología , Mutación , Nucleósido Desaminasas/genética , Nucleósido Desaminasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Regulación hacia Arriba , Replicación Viral
5.
World J Gastroenterol ; 12(46): 7488-96, 2006 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-17167839

RESUMEN

AIM: To investigate the effect of human apolipoprotein B mRNA-editing enzyme catalytic-polypeptide 3G (APOBEC3G) and its N-terminal or C-terminal cytosine deaminase domain-mediated antiviral activity against hepatitis B virus (HBV) in vitro and in vivo. METHODS: The mammalian hepatoma cells HepG2 and HuH7 were cotransfected with APOBEC3G and its N-terminal or C-terminal cytosine deaminase domain expression vector and 1.3-fold-overlength HBV DNA as well as the linear monomeric HBV of genotype B and C. For in vivo study, an HBV vector-based mouse model was used in which APOBEC3G and its N-terminal or C-terminal cytosine deaminase domain expression vectors were co-delivered with 1.3-fold-overlength HBV DNA via high-volume tail vein injection. Levels of hepatitis B virus surface antigen (HBsAg) and hepatitis B virus e antigen (HBeAg) in the media of the transfected cells and in the sera of mice were determined by ELISA. The expression of hepatitis B virus core antigen (HBcAg) in the transfected cells was determined by Western blot analysis. Core-associated HBV DNA was examined by Southern blot analysis. Levels of HBV DNA in the sera of mice as well as HBV core-associated RNA in the liver of mice were determined by quantitative PCR and quantitative RT-PCR analysis, respectively. RESULTS: Human APOBEC3G exerted an anti-HBV activity in a dose-dependent manner in HepG2 cells, and comparable suppressive effects were observed on genotype B and C as that of genotype A. Interestingly, the N-terminal or C-terminal cytosine deaminase domain alone could also inhibit HBV replication in HepG2 cells as well as Huh7 cells. Consistent with in vitro results, the levels of HBsAg in the sera of mice were dramatically decreased, with more than 50 times decrease in the levels of serum HBV DNA and core-associated RNA in the liver of mice treated with APOBEC3G and its N-terminal or C-terminal cytosine deaminase domain as compared to the controls. CONCLUSION: Our findings provide probably the first evidence showing that APOBEC3G and its N-terminal or C-terminal cytosine deaminase domain could suppress HBV replication in vitro and in vivo.


Asunto(s)
Virus de la Hepatitis B/efectos de los fármacos , Nucleósido Desaminasas/química , Nucleósido Desaminasas/farmacología , Proteínas Represoras/química , Proteínas Represoras/farmacología , Replicación Viral/efectos de los fármacos , Desaminasa APOBEC-3G , Animales , Secuencia de Bases , Línea Celular , Citidina Desaminasa , Citosina Desaminasa/química , Citosina Desaminasa/genética , Citosina Desaminasa/farmacología , Replicación del ADN/efectos de los fármacos , ADN Viral/biosíntesis , ADN Viral/genética , Femenino , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos BALB C , Nucleósido Desaminasas/genética , Plásmidos/genética , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Proteínas Represoras/genética , Transfección
6.
J Virol ; 80(23): 11710-22, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16971427

RESUMEN

Cells are categorized as being permissive or nonpermissive according to their ability to produce infectious human immunodeficiency virus type 1 (HIV-1) lacking the viral protein Vif. Nonpermissive cells express the human cytidine deaminase APOBEC3G (hA3G), and Vif has been shown to bind to APOBEC3G and facilitate its degradation. Vif-negative HIV-1 virions produced in nonpermissive cells incorporate hA3G and have a severely reduced ability to produce viral DNA in newly infected cells. While it has been proposed that the reduction in DNA production is due to hA3G-facilitated deamination of cytidine, followed by DNA degradation, we provide evidence here that a decrease in the synthesis of the DNA by reverse transcriptase may account for a significant part of this reduction. During the infection of cells with Vif-negative HIV-1 produced from 293T cells transiently expressing hA3G, much of the inhibition of early (> or =50% reduction) and late (> or =95% reduction) viral DNA production, and of viral infectivity (> or =95% reduction), can occur independently of DNA deamination. The inhibition of the production of early minus-sense strong stop DNA is also correlated with a similar inability of tRNA(3)(Lys) to prime reverse transcription. A similar reduction in tRNA(3)(Lys) priming and viral infectivity is also seen in the naturally nonpermissive cell H9, albeit at significantly lower levels of hA3G expression.


Asunto(s)
Productos del Gen vif/metabolismo , VIH-1/fisiología , Nucleósido Desaminasas/farmacología , Proteínas Represoras/farmacología , Transcripción Reversa/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Desaminasa APOBEC-3G , Línea Celular , Citidina Desaminasa , Regulación de la Expresión Génica , Productos del Gen vif/genética , VIH-1/genética , Humanos , Nucleósido Desaminasas/química , Proteínas Represoras/química , Replicación Viral/fisiología , Productos del Gen vif del Virus de la Inmunodeficiencia Humana
7.
J Virol ; 80(17): 8450-8, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16912295

RESUMEN

The human cytidine deaminases APOBEC3G (hA3G) and APOBEC3F (hA3F) are intracellular antiretroviral factors that can hypermutate nascent reverse transcripts and inhibit the replication of human immunodeficiency virus type 1 (HIV-1). Both enzymes have two cytidine deaminase motifs, although only the C-terminal motif is catalytic. Current models of APOBEC protein function imply editing is the principal mechanism of antiviral activity. In particular, hA3G is a more potent inhibitor of HIV-1 infectivity than hA3F and also induces a greater frequency of mutations in HIV-1 cDNA. We used hA3G/hA3F chimeric proteins to investigate whether cytidine deaminase potential reflects antiviral potency. We show here that the origin of the C-terminal deaminase motif is sufficient to determine the degree of mutation induced in a bacterial assay that measures mutations in chromosomal DNA. In contrast, this was not the case in the context of HIV-1 infection where the N-terminal deaminase motif also modulated the editing capabilities of the chimeras. Surprisingly, although three of the chimeric proteins induced levels of mutation that approximated those of parental hA3F, they displayed lower levels of antiviral activity. Most importantly, real-time PCR experiments revealed that the quantity of reverse transcripts detected in target cells, rather than the mutational burden carried by such DNAs, corresponded closely with viral infectivity. In other words, the antiviral phenotype of APOBEC proteins correlates with their ability to prevent the accumulation of reverse transcripts and not with the induction of hypermutation.


Asunto(s)
Antivirales/farmacología , Citidina Desaminasa/metabolismo , Citosina Desaminasa/metabolismo , ADN Complementario/efectos de los fármacos , VIH-1/efectos de los fármacos , Nucleósido Desaminasas/metabolismo , Proteínas Represoras/metabolismo , Desaminasa APOBEC-3G , Antivirales/metabolismo , Línea Celular , Citosina Desaminasa/genética , Citosina Desaminasa/farmacología , ADN Complementario/metabolismo , ADN Viral/metabolismo , VIH-1/genética , VIH-1/metabolismo , VIH-1/patogenicidad , Humanos , Nucleósido Desaminasas/genética , Nucleósido Desaminasas/farmacología , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Proteínas Represoras/genética , Proteínas Represoras/farmacología
8.
J Virol ; 80(12): 5992-6002, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16731938

RESUMEN

APOBEC3G (APO3G), a cytidine deaminase with two zinc finger domains, inhibits human immunodeficiency virus type 1 replication in the absence of Vif. Here, we provide a comprehensive molecular analysis of the deaminase and nucleic acid binding activities of human APO3G using a pure system containing only one protein component, i.e., highly purified, catalytically active enzyme expressed in a baculovirus system. We demonstrate that APO3G deaminates cytosines in single-stranded DNA (ssDNA) only, whereas it binds efficiently to ssDNA and ssRNA, about half as well to a DNA/RNA hybrid, and poorly to double-stranded DNA and RNA. In addition, the base specificities for deamination and binding of ssDNA are not correlated. The minimum length required for detection of APO3G binding to an ssDNA oligonucleotide in an electrophoretic mobility shift assay is 16 nucleotides. Interestingly, if nucleocapsid protein and APO3G are present in the same reaction, we find that they do not interfere with each other's binding to RNA and a complex containing the RNA and both proteins is formed. Finally, we also identify the functional activities of each zinc finger domain. Thus, although both zinc finger domains have the ability to bind nucleic acids, the first zinc finger contributes more to binding and APO3G encapsidation into virions than finger two. In contrast, deamination is associated exclusively with the second zinc finger. Moreover, zinc finger two is more important than finger one for the antiviral effect, demonstrating a correlation between deaminase and antiviral activities.


Asunto(s)
Nucleósido Desaminasas/farmacología , Proteínas Represoras/farmacología , Desaminasa APOBEC-3G , Antivirales/aislamiento & purificación , Antivirales/metabolismo , Antivirales/farmacología , Baculoviridae/genética , Catálisis , Clonación Molecular , Citidina Desaminasa , ADN de Cadena Simple/metabolismo , Humanos , Nucleósido Desaminasas/aislamiento & purificación , Nucleósido Desaminasas/metabolismo , Oligonucleótidos/metabolismo , Unión Proteica , ARN/metabolismo , Proteínas Represoras/aislamiento & purificación , Proteínas Represoras/metabolismo , Virión/metabolismo , Dedos de Zinc
9.
Br J Cancer ; 88(6): 937-9, 2003 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-12644833

RESUMEN

A recombinant fusion protein of colon carcinoma binding A33 single chain antibody with cytosine deaminase displayed specific antigen binding and enzyme activity in surface plasmon resonance and is catalytic activity assay. In vitro, it selectively increased the toxicity of 5-FC to A33 antigen-positive cells by 300-fold, demonstrating the potency of this ADEPT strategy.


Asunto(s)
Glicoproteínas de Membrana/inmunología , Nucleósido Desaminasas/farmacología , Proteínas Recombinantes de Fusión/farmacología , Antígenos de Neoplasias , Catálisis , Citosina Desaminasa , Escherichia coli/genética , Humanos , Fragmentos de Inmunoglobulinas , Región Variable de Inmunoglobulina , Glicoproteínas de Membrana/farmacología , Nucleósido Desaminasas/química , Nucleósido Desaminasas/inmunología , Reacción en Cadena de la Polimerasa , Células Tumorales Cultivadas
10.
Int J Urol ; 9(10): 567-76, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12445236

RESUMEN

BACKGROUND: The purpose of this study was to investigate the potential of combining cytosine deaminase/5-fluorocytosine (CD/5-FC) gene therapy and radiation therapy (either external beam radiation or radioimmunotherapy [RIT]), for the treatment of prostate cancer. METHODS: Tumor xenografts of CD-transduced LNCaP cells grown in the testes of severe combined immunodeficiency (SCID) mice were used to evaluate antitumor effect. The mice were injected intraperitoneally with 500 mg/kg of 5-FC, or with 5, 15 or 30 mg/kg of 5-fluorouracil (5-FU), for 9 days. The tumors were treated with fractionated radiation at a dose of 1 or 3 Gy/day for 3 days, or I-131 labelled anti-prostate specific antigen (anti-PSA) monoclonal antibody (mAb) administration at a subtherapeutic dose of 20 or 80 micro Ci. Intratumoral and serum concentrations of 5-FU were measured using high performance liquid chromatography. RESULTS: Mice treated with CD/5-FC gene therapy presented a significant tumor growth inhibition comparable to that obtained with 15 mg/kg, 5-FU systemic administration without marked weight loss. Treatment with CD/5-FC gene therapy resulted in higher tumor but lower serum concentrations of 5-FU than treatment with systemic 5-FU chemotherapy. An additive antitumor effect was obtained when CD/5-FC therapy was combined with 1 Gy irradiation, which by itself did not produce a significant antitumor effect. However, the efficacy of CD/5-FC therapy was not enhanced when combined with RIT, probably due to poor accumulation of the mAb as the tumor/blood ratio never exceeded 1. CONCLUSION: These findings indicate that CD/5-FC gene therapy for prostate cancer may function with enhanced antitumor effect when combined with external beam radiation. However, combining CD/5-FC gene therapy and RIT using an anti-PSA mAb may not be effective because of insufficient accumulation of the mAb at the target tumors.


Asunto(s)
Antimetabolitos/farmacología , Flucitosina/farmacología , Terapia Genética/métodos , Nucleósido Desaminasas/farmacología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/terapia , Animales , Terapia Combinada , Citosina Desaminasa , Fluorouracilo/uso terapéutico , Humanos , Masculino , Ratones , Ratones SCID , Modelos Animales , Nucleósido Desaminasas/genética , Neoplasias de la Próstata/radioterapia , Radioinmunoterapia/métodos , Radioterapia/métodos , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...