Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Chem Biol ; 17(12): 1262-1270, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34663942

RESUMEN

DNA deaminase enzymes play key roles in immunity and have recently been harnessed for their biotechnological applications. In base editors (BEs), the combination of DNA deaminase mutator activity with CRISPR-Cas localization confers the powerful ability to directly convert one target DNA base into another. While efforts have been made to improve targeting efficiency and precision, all BEs so far use a constitutively active DNA deaminase. The absence of regulatory control over promiscuous deaminase activity remains a major limitation to accessing the widespread potential of BEs. Here, we reveal sites that permit splitting of DNA cytosine deaminases into two inactive fragments, whose reapproximation reconstitutes activity. These findings allow for the development of split-engineered BEs (seBEs), which newly enable small-molecule control over targeted mutator activity. We show that the seBE strategy facilitates robust regulated editing with BE scaffolds containing diverse deaminases, offering a generalizable solution for temporally controlling precision genome editing.


Asunto(s)
Nucleósido Desaminasas/química , Biotecnología , Sistemas CRISPR-Cas , Citosina/química , ADN/química , Roturas del ADN de Doble Cadena , Escherichia coli , Edición Génica , Conformación de Ácido Nucleico , Nucleósido Desaminasas/genética , Sirolimus/química
2.
Chem Commun (Camb) ; 57(76): 9748-9751, 2021 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-34477187

RESUMEN

Guanosine deaminase (GSDA) in plants specifically deaminates (de)guanosine to produce xanthosine with high specificity, which is further converted to xanthine, a key intermediate in purine metabolism and nitrogen recycling. We solved GSDA's structures from Arabidopsis thaliana in the free and ligand-bound forms at high resolutions. Unlike GDA, the enzyme employs a single-proton shuttle mechanism for catalysis and both the substrate and enzyme undergo structural rearrangements. The last fragment of the enzyme loops back and seals the active site, and the substrate rotates during the reaction, both essential to deamination. We further identified more substrates that could be employed by the enzyme and compare it with other deaminases to reveal the recognition differences of specific substrates. Our studies provide insight into this important enzyme involved in purine metabolism and will potentially aid in the development of deaminase-based gene-editing tools.


Asunto(s)
Arabidopsis/enzimología , Nucleósido Desaminasas/metabolismo , Biocatálisis , Modelos Moleculares , Nucleósido Desaminasas/química
3.
Curr Opin Struct Biol ; 53: 59-68, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29913347

RESUMEN

All messenger RNAs in eukaryotes are modified co-transcriptionally and post-transcriptionally. They are all capped at the 5'-end and polyadenylated at the 3'-end. However, many mRNAs are also found to be chemically modified internally for regulation of mRNA processing, translation, stability, and to recode the message. This review will briefly summarize the structural basis for formation of the two most common modifications found at internal sites in mRNAs; methylation and deamination. The structures of the enzymes that catalyze these modifications show structural similarity to other family members within each modifying enzyme class. RNA methyltransferases, including METTL3/METTL14 responsible for N6-methyladensosine (m6A) formation, share a common structural core and utilize S-adenosyl methionine as a methyl donor. RNA deaminases, including adenosine deaminases acting on RNA (ADARs), also share a common structural core and similar signature sequence motif with conserved residues used for binding zinc and catalyzing the deamination reaction. In spite of recent reports of high resolution structures for members of these two RNA-modifying enzyme families, a great deal remains to be uncovered for a complete understanding of the structural basis for mRNA modification. Of particular interest is the definition of factors that control modification site specificity.


Asunto(s)
Eucariontes/enzimología , Eucariontes/genética , Metiltransferasas/química , Nucleósido Desaminasas/química , ARN Mensajero/metabolismo , Sitios de Unión , Desaminación , Células Eucariotas/metabolismo , Metilación , Modelos Moleculares , Estructura Cuaternaria de Proteína , Procesamiento Postranscripcional del ARN , Proteínas de Unión al ARN/química
4.
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
5.
J Agric Food Chem ; 62(8): 1796-801, 2014 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-24475736

RESUMEN

Brassica oleracea deoxycytidine deaminase (BoDCD), a deoxycytidine deaminase (DCD, EC 3.5.4.14) enzyme, is known to play an important role in the Trichoderma harzianum ETS 323 mediated resistance mechanism in young leaves of B. oleracea var. capitata during Rhizoctonia solani infection. BoDCD potentially neutralizes cytotoxic products of host lipoxygenase activity, and thereby BoDCD restricts the hypersensitivity-related programmed cell death induced in plants during the initial stages of infection. To determine the biochemical characteristics and to partially elucidate the designated functional properties of BoDCD, the enzyme was cloned into an Escherichia coli expression system, and its potential to neutralize the toxic analogues of 2'-deoxycytidine (dC) was examined. BoDCD transformants of E. coli cells were found to be resistant to 2'-deoxycytidine analogues at all of the concentrations tested. The BoDCD enzyme was also overexpressed as a histidine-tagged protein and purified using nickel chelating affinity chromatography. The molecular weight of BoDCD was determined to be 20.8 kDa as visualized by SDS-PAGE. The substrate specificity and other kinetic properties show that BoDCD is more active in neutralizing cytotoxic cytosine ß-d-arabinofuranoside than in deaminating 2'-deoxycytinde to 2'-deoxyuridine in nucleic acids or in metabolizing cytidine to uridine. The optimal temperature and pH of the enzyme were 27 °C and 7.5. The Km and Vmax values of BoDCD were, respectively, 91.3 µM and 1.475 mM for its natural substrate 2'-deoxycytidine and 63 µM and 2.072 mM for cytosine ß-d-arabinofuranoside. The phenomenon of neutralization of cytotoxic dC analogues by BoDCD is discussed in detail on the basis of enzyme biochemical properties.


Asunto(s)
Brassica/enzimología , Nucleósido Desaminasas/química , Proteínas de Plantas/química , Brassica/química , Brassica/genética , Citidina/metabolismo , Citidina Desaminasa , Citosina/metabolismo , Cinética , Peso Molecular , Nucleósido Desaminasas/genética , Nucleósido Desaminasas/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Especificidad por Sustrato , Uridina/metabolismo
6.
J Am Chem Soc ; 135(37): 13927-33, 2013 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-23968233

RESUMEN

A substantial challenge for genomic enzymology is the reliable annotation for proteins of unknown function. Described here is an interrogation of uncharacterized enzymes from the amidohydrolase superfamily using a structure-guided approach that integrates bioinformatics, computational biology, and molecular enzymology. Previously, Tm0936 from Thermotoga maritima was shown to catalyze the deamination of S-adenosylhomocysteine (SAH) to S-inosylhomocysteine (SIH). Homologues of Tm0936 homologues were identified, and substrate profiles were proposed by docking metabolites to modeled enzyme structures. These enzymes were predicted to deaminate analogues of adenosine including SAH, 5'-methylthioadenosine (MTA), adenosine (Ado), and 5'-deoxyadenosine (5'-dAdo). Fifteen of these proteins were purified to homogeneity, and the three-dimensional structures of three proteins were determined by X-ray diffraction methods. Enzyme assays supported the structure-based predictions and identified subgroups of enzymes with the capacity to deaminate various combinations of the adenosine analogues, including the first enzyme (Dvu1825) capable of deaminating 5'-dAdo. One subgroup of proteins, exemplified by Moth1224 from Moorella thermoacetica, deaminates guanine to xanthine, and another subgroup, exemplified by Avi5431 from Agrobacterium vitis S4, deaminates two oxidatively damaged forms of adenine: 2-oxoadenine and 8-oxoadenine. The sequence and structural basis of the observed substrate specificities were proposed, and the substrate profiles for 834 protein sequences were provisionally annotated. The results highlight the power of a multidisciplinary approach for annotating enzymes of unknown function.


Asunto(s)
Nucleósido Desaminasas/química , Dominio Catalítico , Cristalografía por Rayos X , Pruebas de Enzimas , Cinética , Modelos Moleculares , Estructura Molecular , Nucleósido Desaminasas/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato
7.
Anticancer Res ; 33(5): 1899-912, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23645737

RESUMEN

BACKGROUND/AIM: GS 9219 is a double prodrug of antiproliferative nucleotide analog 9-(2-Phosphonylmethoxyethyl)guanine (PMEG), with potent in vivo efficacy against various hematological malignancies. This study investigates the role of adenosine deaminase-like (ADAL) protein in the intracellular activation of GS-9219. MATERIALS AND METHODS: A cell line resistant to 9-(2-Phosphonylmethoxyethyl)-N(6)-cyclopropyl-2,6-diaminopurine (cPrPMEDAP), an intermediate metabolite of GS-9219, was generated and characterized. RESULTS: The resistant cell line was cross-resistant to cPrPMEDAP and GS-9219, due to a defect in the deamination of cPrPMEDAP to PMEG. Mutations in the ADAL gene (H286R and S180N) were identified in the resistant cells that adversely-affected its enzymatic activity. Introduction of the wild-type ADAL gene re-sensitized resistant cells to both cPrPMEDAP and GS-9219. CONCLUSION: The ADAL protein plays an essential role in the intracellular activation of GS-9219 by catalyzing the deamination of cPrPMEDAP metabolite to PMEG. Mutations affecting the activity of ADAL confer resistance to both GS-9219 and its metabolite cPrPMEDAP.


Asunto(s)
Adenina/análogos & derivados , Alanina/análogos & derivados , Resistencia a Antineoplásicos/genética , Mutación/genética , Nucleósido Desaminasas/genética , Purinas/farmacología , Neoplasias del Cuello Uterino/genética , Adenina/farmacología , Alanina/farmacología , Secuencia de Aminoácidos , Antineoplásicos/farmacología , Western Blotting , Femenino , Humanos , Datos de Secuencia Molecular , Nucleósido Desaminasas/química , Nucleósido Desaminasas/metabolismo , Profármacos/farmacología , Conformación Proteica , Homología de Secuencia de Aminoácido , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/tratamiento farmacológico
8.
Angew Chem Int Ed Engl ; 51(44): 11166-9, 2012 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-23038402

RESUMEN

Checking for mistakes: By conjugating a catalytic domain with a guide RNA, deamination activity can be harnessed to repair a specific codon on mRNA. This method can be used for the highly selective repair of point mutations in mRNA by site-selective editing.


Asunto(s)
Nucleósido Desaminasas/química , Mutación Puntual , ARN/química , Adenosina Desaminasa/química , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Proteínas Fluorescentes Verdes/genética , Humanos , Modelos Moleculares , Estructura Molecular , Nucleósido Desaminasas/genética , Nucleósido Desaminasas/metabolismo , ARN/genética , ARN/metabolismo , ARN Mensajero/genética , Proteínas de Unión al ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Biochemistry (Mosc) ; 76(1): 131-46, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21568845

RESUMEN

Enzymatic deamination of bases in DNA or RNA leads to an alteration of flow of genetic information. Adenosine deaminases edit RNA (ADARs, TADs). Specialized cytidine deaminases are involved in RNA/DNA editing in lipid metabolism (APOBEC1) and in innate (APOBEC3 family) and humoral (AID) immunity. APOBEC2 is required for proper muscle development and, along with AID, was implicated in demethylation of DNA. The functions of APOBEC4, APOBEC5, and other deaminases recently discovered by bioinformatics approaches are unknown. What is the basis for the diverse biological functions of enzymes with similar enzyme structure and the same principal enzymatic reaction? AID, APOBEC1, lamprey CDA1, and APOBEC3G enzymes cause uracil DNA glycosylase-dependent induction of mutations when overproduced ectopically in bacteria or yeast. APOBEC2, on the contrary, is nonmutagenic. We studied the effects of the expression of various deaminases in yeast and bacteria. The mutagenic specificities of four deaminases, hAID, rAPOBEC1, hAPOBEC3G, and lamprey CDA1, are strikingly different. This suggests the existence of an intrinsic component of deaminase targeting. The expression of yeast CDD1 and TAD2/TAD3, human APOBEC4, Xanthomonas oryzae APOBEC5, and deaminase encoded by Micromonas sp. gene MICPUN_56782 was nonmutagenic. A lack of a mutagenic effect for Cdd1 is expected because the enzyme functions in the salvage of pyrimidine nucleotides, and it is evolutionarily distant from RNA/DNA editing enzymes. The reason for inactivity of deaminases grouped with APOBEC2 is not obvious from their structures. This can not be explained by protein insolubility and peculiarities of cellular distribution and requires further investigation.


Asunto(s)
Bacterias/enzimología , Proteínas Bacterianas/genética , Proteínas Fúngicas/genética , Mutación , Nucleósido Desaminasas/genética , Levaduras/enzimología , Secuencias de Aminoácidos , Animales , Bacterias/química , Bacterias/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Humanos , Datos de Secuencia Molecular , Nucleósido Desaminasas/química , Nucleósido Desaminasas/metabolismo , Levaduras/química , Levaduras/genética
10.
J Am Chem Soc ; 132(7): 2359-69, 2010 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-20121093

RESUMEN

DesII from Streptomyces venezuelae is a radical SAM (S-adenosyl-l-methionine) enzyme that catalyzes the deamination of TDP-4-amino-4,6-dideoxy-d-glucose to form TDP-3-keto-4,6-dideoxy-d-glucose in the biosynthesis of TDP-d-desosamine. DesII also catalyzes the dehydrogenation of the nonphysiological substrate TDP-D-quinovose to TDP-3-keto-6-deoxy-d-glucose. These properties prompted an investigation of how DesII handles SAM in the redox neutral deamination versus the oxidative dehydrogenation reactions. This work was facilitated by the development of an enzymatic synthesis of TDP-4-amino-4,6-dideoxy-d-glucose that couples a transamination equilibrium to the thermodynamically favorable oxidation of formate. In this study, DesII is found to consume SAM versus TDP-sugar with stoichiometries of 0.96 +/- 0.05 and 1.01 +/- 0.05 in the deamination and dehydrogenation reactions, respectively, using Na(2)S(2)O(4) as the reductant. Importantly, no significant change in stoichiometry is observed when the flavodoxin/flavodoxin NADP(+) oxidoreductase/NADPH reducing system is used in place of Na(2)S(2)O(4). Moreover, there is no evidence of an uncoupled or abortive process in the deamination reaction, as indicated by the observation that dehydrogenation can take place in the absence of an external source of reductant whereas deamination cannot. Mechanistic and biochemical implications of these results are discussed.


Asunto(s)
Proteínas Hierro-Azufre/química , Nucleósido Desaminasas/química , Oxidorreductasas/química , S-Adenosilmetionina/química , Catálisis , Desaminación , Hidrogenación , Proteínas Hierro-Azufre/metabolismo , Cinética , Nucleósido Desaminasas/metabolismo , Oxidación-Reducción , Oxidorreductasas/metabolismo , S-Adenosilmetionina/metabolismo , Streptomyces/enzimología
11.
Biochemistry ; 46(47): 13638-47, 2007 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-17973496

RESUMEN

To estimate the relative importance of alternate routes of spontaneous degradation of DNA and the rate enhancements produced by enzymes catalyzing these reactions, rate constants and thermodynamic activation parameters for the degradation of 2'-deoxynucleosides at 25 degrees C were determined by extrapolation of rates observed in the temperature range between 90 and 200 degrees C in neutral phosphate buffer. Rates of deamination of 2'-deoxycytidine, 1-methylcytosine, and cytidine were found to be identical within experimental error (t1/2 approximately 20 years, 37 degrees C). Rate constants for deamination of 2'-deoxyadenosine and 2'-deoxyguanosine, which could not be determined directly because of rapid glycoside cleavage, were estimated by assuming that methyl replacement should generate reasonable model substrates. The rates of deamination of 9-methyladenine and 9-methylguanine were found to be similar to each other (t1/2 approximately 6000 years, 37 degrees C) and approximately 10(2)-fold slower than the rates of glycoside cleavage in 2'-deoxyadenosine and 2'-deoxyguanosine. The deamination of 2'-deoxyadenosine, 2'-deoxyguanosine, and 2'-deoxycytidine led to accelerated rates of glycoside cleavage. In the exceptional case of 2'-deoxycytidine, deamination and glycoside hydrolysis proceed at very similar rates at all temperatures. Glycoside cleavage proceeds with half-times ranging from 4 years for 2'-deoxyinosine to 40 years for 2'-deoxycytidine (37 degrees C). The rate enhancements produced by DNA glycosylases, estimated by comparison with the rates of these uncatalyzed reactions, are found to be substantially smaller than those produced by deaminases and staphylococcal nuclease.


Asunto(s)
ADN Glicosilasas/química , ADN/metabolismo , Nucleósido Desaminasas/química , Catálisis , ADN/química , ADN Glicosilasas/antagonistas & inhibidores , ADN Glicosilasas/metabolismo , Desaminación , Desoxirribonucleasas/química , Desoxirribonucleasas/metabolismo , Hidrólisis , Cinética , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Nucleósido Desaminasas/antagonistas & inhibidores , Nucleósido Desaminasas/metabolismo , Temperatura , Termodinámica , Thermotoga maritima/enzimología , Thermotoga maritima/fisiología
12.
Nature ; 448(7155): 775-9, 2007 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-17603473

RESUMEN

With many genomes sequenced, a pressing challenge in biology is predicting the function of the proteins that the genes encode. When proteins are unrelated to others of known activity, bioinformatics inference for function becomes problematic. It would thus be useful to interrogate protein structures for function directly. Here, we predict the function of an enzyme of unknown activity, Tm0936 from Thermotoga maritima, by docking high-energy intermediate forms of thousands of candidate metabolites. The docking hit list was dominated by adenine analogues, which appeared to undergo C6-deamination. Four of these, including 5-methylthioadenosine and S-adenosylhomocysteine (SAH), were tested as substrates, and three had substantial catalytic rate constants (10(5) M(-1 )s(-1)). The X-ray crystal structure of the complex between Tm0936 and the product resulting from the deamination of SAH, S-inosylhomocysteine, was determined, and it corresponded closely to the predicted structure. The deaminated products can be further metabolized by T. maritima in a previously uncharacterized SAH degradation pathway. Structure-based docking with high-energy forms of potential substrates may be a useful tool to annotate enzymes for function.


Asunto(s)
Enzimas/química , Enzimas/metabolismo , Relación Estructura-Actividad , Thermotoga maritima/enzimología , Adenina/análogos & derivados , Adenina/metabolismo , Catálisis , Cristalografía por Rayos X , Desaminación , Desoxiadenosinas/metabolismo , Homocisteína/análogos & derivados , Homocisteína/metabolismo , Inosina/análogos & derivados , Inosina/metabolismo , Modelos Moleculares , Nucleósido Desaminasas/química , Nucleósido Desaminasas/metabolismo , Conformación Proteica , S-Adenosilhomocisteína/metabolismo , Especificidad por Sustrato , Tionucleósidos/metabolismo
13.
J Virol ; 81(15): 8236-46, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17522211

RESUMEN

Human immunodeficiency virus type 1 (HIV-1) Vif counteracts the antiviral activity of the human cytidine deaminase APOBEC3G (APO3G) by inhibiting its incorporation into virions. This has been attributed to the Vif-induced degradation of APO3G by cytoplasmic proteasomes. We recently demonstrated that although APO3G has a natural tendency to form RNA-dependent homo-multimers, multimerization was not essential for encapsidation into HIV-1 virions or antiviral activity. We now demonstrate that a multimerization-defective APO3G variant (APO3G C97A) is able to assemble into RNase-sensitive high-molecular-mass (HMM) complexes, suggesting that homo-multimerization of APO3G and assembly into HMM complexes are unrelated RNA-dependent processes. Interestingly, APO3G C97A was highly resistant to Vif-induced degradation even though the two proteins were found to interact in coimmunoprecipitation experiments and exhibited partial colocalization in transfected HeLa cells. Surprisingly, encapsidation and antiviral activity of APO3G C97A were both inhibited by Vif despite resistance to degradation. These results demonstrate that targeting of APO3G to proteasome degradation and interference with viral encapsidation are distinct functional properties of Vif.


Asunto(s)
Productos del Gen vif/metabolismo , VIH-1/metabolismo , Nucleósido Desaminasas/metabolismo , Proteínas Represoras/metabolismo , Desaminasa APOBEC-3G , Animales , Citidina Desaminasa , Productos del Gen vif/genética , VIH-1/patogenicidad , Células HeLa , Humanos , Mutación , Nucleósido Desaminasas/química , Nucleósido Desaminasas/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas Represoras/química , Proteínas Represoras/genética , Virión , Productos del Gen vif del Virus de la Inmunodeficiencia Humana
14.
Virology ; 364(2): 486-93, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17434555

RESUMEN

The mammalian APOBEC3 proteins are cytidine deaminases that function as inhibitors of retrovirus replication and retrotransposon mobility. An issue that has remained controversial is whether the editing of deoxycytidine residues to deoxyuridine is necessary and sufficient for this inhibition or whether APOBEC3 proteins also exert a second, distinct inhibitory mechanism. Here, we present an analysis of the ability of mutants of APOBEC3G and APOBEC3B, both of which contain two consensus cytidine deaminase active sites, to inhibit the replication of human immunodeficiency virus. Our data confirm that APOBEC3G only contains a single, carboxy-terminal active site but, surprisingly, reveal that both cytidine deaminase consensus sequences in APOBEC3B are enzymatically active. Enzymatically inactive mutant forms of APOBEC3G and APOBEC3B were found to retain the ability to inhibit the infectivity of HIV-1 virions produced in their presence by approximately 4-fold and approximately 8-fold, respectively. While this inhibition was significantly less than the level seen with wild-type forms of A3G or A3B, these data, nevertheless argue that the inhibition of HIV-1 by APOBEC3 proteins is at least partly independent of DNA editing.


Asunto(s)
Citidina Desaminasa/química , Citidina Desaminasa/fisiología , VIH-1/fisiología , Desaminasa APOBEC-3G , Sustitución de Aminoácidos , Antirretrovirales/química , Antirretrovirales/metabolismo , Línea Celular , Citidina Desaminasa/genética , VIH-1/genética , Humanos , Técnicas In Vitro , Antígenos de Histocompatibilidad Menor , Mutagénesis Sitio-Dirigida , Nucleósido Desaminasas/química , Nucleósido Desaminasas/genética , Nucleósido Desaminasas/fisiología , Estructura Terciaria de Proteína , Edición de ARN , ARN Viral/genética , ARN Viral/metabolismo , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas Represoras/fisiología
15.
J Virol ; 81(8): 3807-15, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17267497

RESUMEN

The human immunodeficiency virus type-1 (HIV-1) accessory protein Vif serves to neutralize the human antiviral proteins apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3G (APOBEC3G [A3G]) and A3F. As such, the therapeutic blockade of Vif function represents a logical objective for rational drug design. To facilitate such endeavors, we have employed molecular genetics to define features of A3G that are required for its interaction with Vif. Using alanine-scanning mutations and multiple different substitutions at key residues, we confirm the central role played by the aspartic acid at position 128 and identify proline 129 and aspartic acid 130 as important contributory residues. The overall negative charge of this 3-amino-acid motif appears critical for recognition by Vif, as single lysine substitutions are particularly deleterious and a double alanine substitution at positions 128 and 130 is far more inhibitory than single-residue mutations at either position. Our analyses also reveal that the immediately adjacent 4 amino acids, residues 124 to 127, are important for the packaging of A3G into HIV-1 particles. Most important are tyrosine 124 and tryptophan 127, and mutations at these positions can ablate virion incorporation, as well as the capacity to inhibit virus infection. Thus, while pharmacologic agents that target the acidic motif at residues 128 to 130 have the potential to rescue A3G expression by occluding recognition by Vif, care will have to be taken not to perturb the contributions of the neighboring 124-to-127 region to packaging if such agents are to have therapeutic benefit by promoting A3G incorporation into progeny virions.


Asunto(s)
Productos del Gen vif/metabolismo , VIH-1/fisiología , Nucleósido Desaminasas/química , Nucleósido Desaminasas/metabolismo , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Ensamble de Virus , Desaminasa APOBEC-3G , Secuencias de Aminoácidos , Sustitución de Aminoácidos , Línea Celular , Citidina Desaminasa , Humanos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Nucleósido Desaminasas/genética , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteínas Represoras/genética , Virión/fisiología , Productos del Gen vif del Virus de la Inmunodeficiencia Humana
16.
J Biol Chem ; 282(3): 1585-94, 2007 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-17142455

RESUMEN

APOBEC3G and APOBEC3F are cytidine deaminase with duplicative cytidine deaminase motifs that restrict HIV-1 replication by catalyzing C-to-U transitions on nascent viral cDNA. Despite 60% protein sequence similarity, APOBEC3F and APOBEC3G have a different target consensus sequence for editing, and importantly, APOBEC3G has 10-fold higher anti-HIV activity than APOBEC3F. Thus, APOBEC3F and APOBEC3G may have distinctive characteristics that account for their functional differences. Here, we have biochemically characterized human APOBEC3F and APOBEC3G protein complexes as a function of the HIV-1 life cycle. APOBEC3G was previously shown to form RNase-sensitive, enzymatically inactive, high molecular mass complexes in immortalized cells, which are converted into enzymatically active, low molecular mass complexes by RNase digestion. We found that APOBEC3F also formed high molecular mass complexes in these cells, but these complexes were resistant to RNase treatment. Further, the N-terminal half determined RNase sensitivity and was necessary for the high molecular mass complex assembly of APOBEC3G but not APOBEC3F. Unlike APOBEC3F, APOBEC3G strongly interacted with cellular proteins via disulfide bonds. Inside virions, both APOBEC3F and APOBEC3G were found in viral cores, but APOBEC3G was associated with low molecular mass, whereas APOBEC3F was still retained in high molecular mass complexes. After cell entry, both APOBEC3F and APOBEC3G were localized in low molecular mass complexes associated with viral reverse transcriptional machinery. These results demonstrate that APOBEC3F and APOBEC3G complexes undergo dynamic conversion during HIV-1 infection and also reveal biochemical differences that likely determine their different anti-HIV-1 activity.


Asunto(s)
Citosina Desaminasa/fisiología , Regulación de la Expresión Génica , Nucleósido Desaminasas/fisiología , Proteínas Represoras/fisiología , Desaminasa APOBEC-3G , Secuencias de Aminoácidos , Fármacos Anti-VIH/química , Línea Celular , Citidina Desaminasa/química , Citosina Desaminasa/química , ADN Complementario/metabolismo , ADN Viral/química , Disulfuros/química , VIH-1/metabolismo , Humanos , Nucleósido Desaminasas/química , Plásmidos/metabolismo , Proteínas Represoras/química , Ribonucleasas/metabolismo , Transcripción Genética
17.
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
18.
J Biol Chem ; 281(50): 38122-6, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17079235

RESUMEN

Human APOBEC3G (hA3G) is a cytidine deaminase that restricts human immunodeficiency virus (HIV)-1 infection in a vif (the virion infectivity factor from HIV)-dependent manner. hA3G from HIV-permissive activated CD4+ T-cells exists as an inactive, high molecular mass (HMM) complex that can be transformed in vitro into an active, low molecular mass (LMM) variant comparable with that of HIV-non-permissive CD4+ T-cells. Here we present low resolution structures of hA3G in HMM and LMM forms determined by small angle x-ray scattering and advanced shape reconstruction methods. The results show that LMM particles have an extended shape, dissimilar to known cytidine deaminases, featuring novel tail-to-tail dimerization. Shape analysis of LMM and HMM structures revealed how symmetric association of dimers could lead to minimal HMM variants. These observations imply that the disruption of cellular HMM particles may require regulation of protein-RNA, as well as protein-protein interactions, which has implications for therapeutic development.


Asunto(s)
Nucleósido Desaminasas/metabolismo , Proteínas Represoras/metabolismo , Desaminasa APOBEC-3G , Linfocitos T CD4-Positivos/metabolismo , Línea Celular , Citidina Desaminasa , Dimerización , Humanos , Peso Molecular , Nanoestructuras , Nucleósido Desaminasas/química , Proteínas Represoras/química , Dispersión de Radiación
19.
Proc Natl Acad Sci U S A ; 103(42): 15588-93, 2006 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-17030807

RESUMEN

APOBEC3G (A3G) and related deoxycytidine deaminases are potent intrinsic antiretroviral factors. A3G is expressed either as an enzymatically active low-molecular-mass (LMM) form or as an enzymatically inactive high-molecular-mass (HMM) ribonucleoprotein complex. Resting CD4 T cells exclusively express LMM A3G, where it functions as a powerful postentry restriction factor for HIV-1. Activation of CD4 T cells promotes the recruitment of LMM A3G into 5- to 15-MDa HMM complexes whose function is unknown. Using tandem affinity purification techniques coupled with MS, we identified Staufen-containing RNA-transporting granules and Ro ribonucleoprotein complexes as specific components of HMM A3G complexes. Analysis of RNAs in these complexes revealed Alu and small Y RNAs, two of the most prominent nonautonomous mobile genetic elements in human cells. These retroelement RNAs are recruited into Staufen-containing RNA-transporting granules in the presence of A3G. Retrotransposition of Alu and hY RNAs depends on the reverse transcriptase machinery provided by long interspersed nucleotide elements 1 (L1). We now show that A3G greatly inhibits L1-dependent retrotransposition of marked Alu retroelements not by inhibiting L1 function but by sequestering Alu RNAs in cytoplasmic HMM A3G complexes away from the nuclear L1 enzymatic machinery. These findings identify nonautonomous Alu and hY retroelements as natural cellular targets of A3G and highlight how different forms of A3G uniquely protect cells from the threats posed by exogenous retroviruses (LMM A3G) and endogenous retroelements (HMM A3G).


Asunto(s)
Elementos Alu , Mutagénesis Insercional , Nucleósido Desaminasas/metabolismo , Proteínas Represoras/metabolismo , Desaminasa APOBEC-3G , Línea Celular , Citidina Desaminasa , VIH-1/genética , VIH-1/metabolismo , Humanos , Sustancias Macromoleculares , Conformación de Ácido Nucleico , Nucleósido Desaminasas/química , ARN/genética , ARN/metabolismo , Proteínas Represoras/química , Ribonucleoproteínas/metabolismo
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...