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










Base de datos
Intervalo de año de publicación
2.
Antiviral Res ; 218: 105716, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37690700

RESUMEN

Sangivamycin (S) is an adenosine (A) nucleoside analog with low nanomolar antiviral activity against SARS-CoV-2 in vitro. Previously, low nanomolar antiviral efficacy was revealed when tested against multiple viral variants in several cell types. SARS-CoV-2 RNA isolated from live virus infected cells and the virions released from these cells was analyzed by mass spectrometry (MS) for S incorporation. Dose-dependent incorporation occurred up to 1.8 S per 1,000 nucleotides (49 S per genome) throughout the viral genomes isolated from both infected cells and viral particles, but this incorporation did not change the viral mutation rate. In contrast, host mRNA, affinity purified from the same infected and treated cells, contained little or no S. Sangivamycin triphosphate (STP) was synthesized to evaluate its incorporation into RNA by recombinant SARS-CoV-2 RNA-dependent RNA polymerase (RdRp) under defined in vitro conditions. SARS-CoV-2 RdRp showed that S was not a chain terminator and S containing oligonucleotides templated as A. Though the antiviral mechanism remains to be determined, the data suggests that SARS-CoV-2 RdRp incorporates STP into SARS-CoV-2 RNA, which does not significantly impair viral RNA synthesis or the mutation rate.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , ARN Viral/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Antivirales/química
3.
JCI Insight ; 7(1)2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34807849

RESUMEN

Sangivamycin is a nucleoside analog that is well tolerated by humans and broadly active against phylogenetically distinct viruses, including arenaviruses, filoviruses, and orthopoxviruses. Here, we show that sangivamycin is a potent antiviral against multiple variants of replicative severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) with half-maximal inhibitory concentration in the nanomolar range in several cell types. Sangivamycin suppressed SARS-CoV-2 replication with greater efficacy than remdesivir (another broad-spectrum nucleoside analog). When we investigated sangivamycin's potential for clinical administration, pharmacokinetic; absorption, distribution, metabolism, and excretion (ADME); and toxicity properties were found to be favorable. When tested in combination with remdesivir, efficacy was additive rather than competitive against SARS-CoV-2. The proven safety in humans, long half-life, potent antiviral activity (compared to remdesivir), and combinatorial potential suggest that sangivamycin is likely to be efficacious alone or in combination therapy to suppress viremia in patients. Sangivamycin may also have the ability to help combat drug-resistant or vaccine-escaping SARS-CoV-2 variants since it is antivirally active against several tested variants. Our results support the pursuit of sangivamycin for further preclinical and clinical development as a potential coronavirus disease 2019 therapeutic.


Asunto(s)
Antivirales , Nucleósidos de Pirimidina , SARS-CoV-2/efectos de los fármacos , Animales , Antivirales/farmacocinética , Antivirales/farmacología , Antivirales/toxicidad , COVID-19/virología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Femenino , Humanos , Masculino , Ratones , Nucleósidos de Pirimidina/farmacocinética , Nucleósidos de Pirimidina/farmacología , Nucleósidos de Pirimidina/toxicidad , Células Vero
4.
J Biol Chem ; 297(6): 101390, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34767799

RESUMEN

RNA represents a potential target for new antiviral therapies, which are urgently needed to address public health threats such as the human immunodeficiency virus (HIV). We showed previously that the interaction between the viral Tat protein and the HIV-1 trans-activation response (TAR) RNA was blocked by TB-CP-6.9a. This cyclic peptide was derived from a TAR-binding loop that emerged during lab evolution of a TAR-binding protein (TBP) family. Here we synthesized and characterized a next-generation, cyclic-peptide library based on the TBP scaffold. We sought to identify conserved RNA-binding interactions and the influence of cyclization linkers on RNA binding and antiviral activity. A diverse group of cyclization linkers, encompassing disulfide bonds to bicyclic aromatic staples, was used to restrain the cyclic peptide geometry. Thermodynamic profiling revealed specific arginine-rich sequences with low to submicromolar affinity driven by enthalpic and entropic contributions. The best compounds exhibited no appreciable off-target binding to related molecules, such as BIV TAR and human 7SK RNAs. A specific arginine-to-lysine change in the highest affinity cyclic peptide reduced TAR binding by tenfold, suggesting that TBP-derived cyclic peptides use an arginine-fork motif to recognize the TAR major groove while differentiating the mode of binding from other TAR-targeting molecules. Finally, we showed that HIV infectivity in cell culture was reduced in the presence of cyclic peptides constrained by methylene or naphthalene-based linkers. Our findings provide insight into the molecular determinants required for HIV-1 TAR recognition and antiviral activity. These findings are broadly relevant to the development of antivirals that target RNA molecules.


Asunto(s)
Antivirales/química , VIH-1/química , Péptidos Cíclicos/química , ARN Viral/química , Células HEK293 , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , VIH-1/genética , VIH-1/metabolismo , Humanos , Unión Proteica , ARN Viral/genética , ARN Viral/metabolismo
5.
Viruses ; 13(1)2020 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-33396288

RESUMEN

Filoviruses, such as Ebola virus and Marburg virus, are of significant human health concern. From 2013 to 2016, Ebola virus caused 11,323 fatalities in Western Africa. Since 2018, two Ebola virus disease outbreaks in the Democratic Republic of the Congo resulted in 2354 fatalities. Although there is progress in medical countermeasure (MCM) development (in particular, vaccines and antibody-based therapeutics), the need for efficacious small-molecule therapeutics remains unmet. Here we describe a novel high-throughput screening assay to identify inhibitors of Ebola virus VP40 matrix protein association with viral particle assembly sites on the interior of the host cell plasma membrane. Using this assay, we screened nearly 3000 small molecules and identified several molecules with the desired inhibitory properties. In secondary assays, one identified compound, sangivamycin, inhibited not only Ebola viral infectivity but also that of other viruses. This finding indicates that it is possible for this new VP40-based screening method to identify highly potent MCMs against Ebola virus and its relatives.


Asunto(s)
Antivirales/farmacología , Ebolavirus/efectos de los fármacos , Nucleoproteínas/antagonistas & inhibidores , Proteínas del Núcleo Viral/antagonistas & inhibidores , Animales , Antivirales/química , Antivirales/uso terapéutico , Chlorocebus aethiops , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Ebolavirus/genética , Regulación Viral de la Expresión Génica/efectos de los fármacos , Células HEK293 , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebre Hemorrágica Ebola/virología , Humanos , Contramedidas Médicas , Estructura Molecular , Nucleoproteínas/química , Nucleósidos de Pirimidina/farmacología , Células Vero , Proteínas del Núcleo Viral/química , Liberación del Virus/efectos de los fármacos
6.
Subcell Biochem ; 93: 193-219, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31939152

RESUMEN

The DNA mutagenic enzyme known as APOBEC3G (A3G) plays a critical role in innate immunity to Human Immunodeficiency Virus-1 (HIV-1 ). A3G is a zinc-dependent enzyme that mutates select deoxycytidines (dC) to deoxyuridine (dU) through deamination within nascent single stranded DNA (ssDNA) during HIV reverse transcription. This activity requires that the enzyme be delivered to viral replication complexes by redistributing from the cytoplasm of infected cells to budding virions through what appears to be an RNA-dependent process. Once inside infected cells, A3G must bind to nascent ssDNA reverse transcripts for dC to dU base modification gene editing. In this chapter we will discuss data indicating that ssDNA deaminase activity of A3G is regulated by RNA binding to A3G and ribonucleoprotein complex formation along with evidence suggesting that RNA-selective interactions with A3G are temporally and mechanistically important in this process.


Asunto(s)
Desaminasa APOBEC-3G/metabolismo , VIH-1/inmunología , Inmunidad Innata , Ribonucleoproteínas/metabolismo , Humanos
7.
Trends Mol Med ; 24(5): 507-520, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29609878

RESUMEN

The infectivity of HIV depends on overcoming APOBEC3 (A3) innate immunity, predominantly through the expression of the viral protein Vif, which induces A3 degradation in the proteasome. Disruption of the functional interactions of Vif enables A3 mutagenesis of the HIV genome during viral replication, which can result in a broadly neutralizing antiviral effect. Vif function requires self-association along with interactions with A3 proteins, protein chaperones, and factors of the ubiquitination machinery and these are described here as a potential platform for novel antiviral drug discovery. This Review will examine the current state of development of Vif inhibitors that we believe to have therapeutic and functional cure potential.


Asunto(s)
Antirretrovirales/uso terapéutico , Citosina Desaminasa/inmunología , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/inmunología , Desaminasas APOBEC , Citidina Desaminasa , Citosina Desaminasa/metabolismo , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/inmunología , VIH-1/fisiología , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Humanos , Unión Proteica/efectos de los fármacos , Proteolisis/efectos de los fármacos , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo
8.
ACS Chem Biol ; 12(6): 1674-1682, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28448121

RESUMEN

The HIV-1 frameshift-stimulating (FSS) RNA, a regulatory RNA of critical importance in the virus' life cycle, has been posited as a novel target for anti-HIV drug development. We report the synthesis and evaluation of triazole-containing compounds able to bind the FSS with high affinity and selectivity. Readily accessible synthetically, these compounds are less toxic than previously reported olefin congeners. We show for the first time that FSS-targeting compounds have antiviral activity against replication-competent HIV in human cells, including a highly cytopathic, multidrug-resistant strain. These results support the viability of the HIV-1 FSS RNA as a therapeutic target and more generally highlight opportunities for synthetic molecule-mediated interference with protein recoding in a wide range of organisms.


Asunto(s)
Sistema de Lectura Ribosómico/efectos de los fármacos , VIH-1/genética , ARN Viral/efectos de los fármacos , Triazoles/farmacología , Replicación Viral/efectos de los fármacos , Fármacos Anti-VIH/química , Fármacos Anti-VIH/farmacología , Línea Celular , VIH-1/efectos de los fármacos , Humanos , Terapia Molecular Dirigida , ARN Viral/genética
9.
J Biol Chem ; 292(21): 8642-8656, 2017 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-28381554

RESUMEN

APOBEC3G (A3G) belongs to the AID/APOBEC protein family of cytidine deaminases (CDA) that bind to nucleic acids. A3G mutates the HIV genome by deamination of dC to dU, leading to accumulation of virus-inactivating mutations. Binding to cellular RNAs inhibits A3G binding to substrate single-stranded (ss) DNA and CDA activity. Bulk RNA and substrate ssDNA bind to the same three A3G tryptic peptides (amino acids 181-194, 314-320, and 345-374) that form parts of a continuously exposed protein surface extending from the catalytic domain in the C terminus of A3G to its N terminus. We show here that the A3G tyrosines 181 and 315 directly cross-linked ssDNA. Binding experiments showed that a Y315A mutation alone significantly reduced A3G binding to both ssDNA and RNA, whereas Y181A and Y182A mutations only moderately affected A3G nucleic acid binding. Consistent with these findings, the Y315A mutant exhibited little to no deaminase activity in an Escherichia coli DNA mutator reporter, whereas Y181A and Y182A mutants retained ∼50% of wild-type A3G activity. The Y315A mutant also showed a markedly reduced ability to assemble into viral particles and had reduced antiviral activity. In uninfected cells, the impaired RNA-binding capacity of Y315A was evident by a shift of A3G from high-molecular-mass ribonucleoprotein complexes to low-molecular-mass complexes. We conclude that Tyr-315 is essential for coordinating ssDNA interaction with or entry to the deaminase domain and hypothesize that RNA bound to Tyr-315 may be sufficient to competitively inhibit ssDNA deaminase-dependent antiviral activity.


Asunto(s)
Desaminasa APOBEC-3G/metabolismo , ADN de Cadena Simple/metabolismo , ADN Viral/metabolismo , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Mutagénesis , ARN Viral/metabolismo , Desaminasa APOBEC-3G/química , Desaminasa APOBEC-3G/genética , Sustitución de Aminoácidos , Línea Celular , ADN de Cadena Simple/química , ADN de Cadena Simple/genética , ADN Viral/química , ADN Viral/genética , Infecciones por VIH/genética , VIH-1/química , VIH-1/genética , Humanos , Mutación Missense , Dominios Proteicos , ARN Viral/química , ARN Viral/genética , Tirosina/química , Tirosina/genética , Tirosina/metabolismo
10.
Antiviral Res ; 136: 51-59, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27825797

RESUMEN

Camptothecin (CPT) is a natural product discovered to be active against various cancers through its ability to inhibit Topoisomerase I (TOP1). CPT analogs also have anti-HIV-1 (HIV) activity that was previously shown to be independent of TOP1 inhibition. We show that a cancer inactive CPT analog (O2-16) inhibits HIV infection by disrupting multimerization of the HIV protein Vif. Antiviral activity depended on the expression of the cellular viral restriction factor APOBEC3G (A3G) that, in the absence of functional Vif, has the ability to hypermutate HIV proviral DNA during reverse transcription. Our studies demonstrate that O2-16 has low cytotoxicity and inhibits Vif-dependent A3G degradation, enabling A3G packaging into HIV viral particles that results in A3G signature hypermutations in viral genomes. This antiviral activity was A3G-dependent and broadly neutralizing against sixteen HIV clinical isolates from groups M (subtypes A-G), N, and O as well as seven single and multi-drug resistant strains of HIV. Molecular modeling predicted binding near the PPLP motif crucial for Vif multimerization and activity. O2-16 also was active in blocking Vif degradation of APOBEC3F (A3F). We propose that CPT analogs not active against TOP1 have novel therapeutic potential as Vif antagonists that enable A3G-dependent hypermutation of HIV.


Asunto(s)
Desaminasa APOBEC-3G/metabolismo , Camptotecina/análogos & derivados , ADN-Topoisomerasas de Tipo I/metabolismo , VIH-1/efectos de los fármacos , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo , Desaminasa APOBEC-3G/genética , Camptotecina/farmacología , Línea Celular , Farmacorresistencia Viral/genética , Genoma Viral , Infecciones por VIH/virología , VIH-1/genética , VIH-1/fisiología , Humanos , Modelos Moleculares , Mutación , Unión Proteica , Multimerización de Proteína/efectos de los fármacos , Virión/metabolismo , Replicación Viral , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/química
11.
Trends Biochem Sci ; 41(7): 578-594, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27283515

RESUMEN

The APOBEC (apolipoprotein B mRNA editing catalytic polypeptide-like) family of proteins have diverse and important functions in human health and disease. These proteins have an intrinsic ability to bind to both RNA and single-stranded (ss) DNA. Both function and tissue-specific expression varies widely for each APOBEC protein. We are beginning to understand that the activity of APOBEC proteins is regulated through genetic alterations, changes in their transcription and mRNA processing, and through their interactions with other macromolecules in the cell. Loss of cellular control of APOBEC activities leads to DNA hypermutation and promiscuous RNA editing associated with the development of cancer or viral drug resistance, underscoring the importance of understanding how APOBEC proteins are regulated.


Asunto(s)
Desaminasas APOBEC/química , Desaminasas APOBEC/metabolismo , Humanos
12.
Methods ; 107: 10-22, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-26988126

RESUMEN

There are eleven members in the human APOBEC family of proteins that are evolutionarily related through their zinc-dependent cytidine deaminase domains. The human APOBEC gene clusters arose on chromosome 6 and 22 through gene duplication and divergence to where current day APOBEC proteins are functionally diverse and broadly expressed in tissues. APOBEC serve enzymatic and non enzymatic functions in cells. In both cases, formation of higher-order structures driven by APOBEC protein-protein interactions and binding to RNA and/or single stranded DNA are integral to their function. In some circumstances, these interactions are regulatory and modulate APOBEC activities. We are just beginning to understand how macromolecular interactions drive processes such as APOBEC subcellular compartmentalization, formation of holoenzyme complexes, gene targeting, foreign DNA restriction, anti-retroviral activity, formation of ribonucleoprotein particles and APOBEC degradation. Protein-protein and protein-nucleic acid cross-linking methods coupled with mass spectrometry, electrophoretic mobility shift assays, glycerol gradient sedimentation, fluorescence anisotropy and APOBEC deaminase assays are enabling mapping of interacting surfaces that are essential for these functions. The goal of this methods review is through example of our research on APOBEC3G, describe the application of cross-linking methods to characterize and quantify macromolecular interactions and their functional implications. Given the homology in structure and function, it is proposed that these methods will be generally applicable to the discovery process for other APOBEC and RNA and DNA editing and modifying proteins.


Asunto(s)
Desaminasa APOBEC-3G/química , Complejos Multiproteicos/química , Mapeo de Interacción de Proteínas/métodos , Desaminasa APOBEC-3G/genética , Citidina Desaminasa/química , Citidina Desaminasa/genética , Humanos , Familia de Multigenes , Complejos Multiproteicos/genética , Conformación Proteica , Edición de ARN/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética
13.
ACS Chem Biol ; 11(1): 88-94, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26496521

RESUMEN

Human Immunodeficiency Virus (HIV) type 1 uses a -1 programmed ribosomal frameshift (-1 PRF) event to translate its enzymes from the same transcript used to encode the virus' structural proteins. The frequency of this event is highly regulated, and significant deviation from the normal 5-10% frequency has been demonstrated to decrease viral infectivity. Frameshifting is primarily regulated by the Frameshift Stimulatory Signal RNA (FSS-RNA), a thermodynamically stable, highly conserved stem loop that has been proposed as a therapeutic target. We describe the design, synthesis, and testing of a series of N-methyl peptides able to bind the HIV-1 FSS RNA stem loop with low nanomolar affinity and high selectivity. Surface plasmon resonance (SPR) data indicates increased affinity is a reflection of a substantially enhanced on rate. Compounds readily penetrate cell membranes and inhibit HIV infectivity in a pseudotyped virus assay. Viral infectivity inhibition correlates with compound-dependent changes in the ratios of Gag and Gag-Pol in virus particles. As the first compounds with both single digit nanomolar affinities for the FSS RNA and an ability to inhibit HIV in cells, these studies support the use of N-methylation for enhancing the affinity, selectivity, and bioactivity of RNA-binding peptides.


Asunto(s)
Sistema de Lectura Ribosómico/fisiología , VIH-1/química , ARN Viral/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Secuencia de Bases , Sistema de Lectura Ribosómico/genética , Células HEK293 , VIH-1/efectos de los fármacos , Humanos , Metilación , Datos de Secuencia Molecular , Estructura Molecular , Péptidos/síntesis química , Péptidos/metabolismo , Unión Proteica , ARN Viral/química
14.
Wiley Interdiscip Rev RNA ; 5(4): 493-508, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24664896

RESUMEN

Cytidine deaminases have important roles in the regulation of nucleoside/deoxynucleoside pools for DNA and RNA synthesis. The APOBEC family of cytidine deaminases (named after the first member of the family that was described, Apolipoprotein B mRNA Editing Catalytic Subunit 1, also known as APOBEC1 or A1) is a fascinating group of mutagenic proteins that use RNA and single-stranded DNA (ssDNA) as substrates for their cytidine or deoxycytidine deaminase activities. APOBEC proteins and base-modification nucleic acid editing have been the subject of numerous publications, reviews, and speculation. These proteins play diverse roles in host cell defense, protecting cells from invading genetic material, enabling the acquired immune response to antigens and changing protein expression at the level of the genetic code in mRNA or DNA. The amazing power these proteins have for interphase cell functions relies on structural and biochemical properties that are beginning to be understood. At the same time, the substrate selectivity of each member in the family and their regulation remains to be elucidated. This review of the APOBEC family will focus on an open question in regulation, namely what role the interactions of these proteins with RNA have in editing substrate recognition or allosteric regulation of DNA mutagenic and host-defense activities.


Asunto(s)
Citidina Desaminasa/metabolismo , ARN/metabolismo , Desaminasas APOBEC-1 , Sitios de Unión
15.
J Med Chem ; 57(3): 723-32, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24387306

RESUMEN

The life cycle of the human immunodeficiency virus type 1 (HIV-1) has an absolute requirement for ribosomal frameshifting during protein translation in order to produce the polyprotein precursor of the viral enzymes. While an RNA stem-loop structure (the "HIV-1 Frameshift Stimulating Signal", or HIV-1 FSS) controls the frameshift efficiency and has been hypothesized as an attractive therapeutic target, developing compounds that selectively bind this RNA and interfere with HIV-1 replication has proven challenging. Building on our prior discovery of a "hit" molecule able to bind this stem-loop, we now report the development of compounds displaying high affinity for the HIV-1 FSS. These compounds are able to enhance frameshifting more than 50% in a dual-luciferase assay in human embryonic kidney cells, and they strongly inhibit the infectivity of pseudotyped HIV-1 virions.


Asunto(s)
Fármacos Anti-VIH/farmacología , VIH-1/genética , Quinolinas/farmacología , ARN Viral/genética , Fármacos Anti-VIH/síntesis química , Fármacos Anti-VIH/química , Permeabilidad de la Membrana Celular , Supervivencia Celular/efectos de los fármacos , Disulfuros/síntesis química , Disulfuros/química , Disulfuros/farmacología , Sistema de Lectura Ribosómico , Células HEK293 , VIH-1/efectos de los fármacos , VIH-1/patogenicidad , Humanos , Luciferasas de Luciérnaga/genética , Luciferasas de Luciérnaga/metabolismo , Luciferasas de Renilla/genética , Luciferasas de Renilla/metabolismo , Conformación de Ácido Nucleico , Quinolinas/síntesis química , Quinolinas/química , Estereoisomerismo , Virión/efectos de los fármacos , Virión/patogenicidad , Replicación Viral/efectos de los fármacos
16.
Semin Cell Dev Biol ; 23(3): 258-68, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22001110

RESUMEN

APOBEC1 is a cytidine deaminase that edits messenger RNAs and was the first enzyme in the APOBEC family to be functionally characterized. Under appropriate conditions APOBEC1 also deaminates deoxycytidine in single-stranded DNA (ssDNA). The other ten members of the APOBEC family have not been fully characterized however several have deoxycytidine deaminase activity on ssDNAs. Despite the nucleic acid substrate preferences of different APOBEC proteins, a common feature appears to be their intrinsic ability to bind to RNA as well as to ssDNA. RNA binding to APOBEC proteins together with protein-protein interactions, post-translation modifications and subcellular localization serve as biological modulators controlling the DNA mutagenic activity of these potentially genotoxic proteins.


Asunto(s)
Citidina Desaminasa/metabolismo , Animales , Citidina Desaminasa/química , ADN de Cadena Simple/metabolismo , Humanos , ARN/metabolismo , Edición de ARN
17.
J Biol Chem ; 283(48): 33329-36, 2008 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-18842592

RESUMEN

Human APOBEC3G (hA3G) is a cytidine deaminase active on HIV single-stranded DNA. Small angle x-ray scattering and molecular envelope restorations predicted a C-terminal dimeric model for RNA-depleted hA3G in solution. Each subunit was elongated, suggesting that individual domains of hA3G are solvent-exposed and therefore may interact with other macromolecules even as isolated substructures. In this study, co-immunoprecipitation and in-cell quenched fluorescence resonance energy transfer assays reveal that hA3G forms RNA-independent oligomers through interactions within its C terminus. Residues 209-336 were necessary and sufficient for homoligomerization. N-terminal domains of hA3G were unable to multimerize but remained functional for Gag and viral infectivity factor (Vif) interactions when expressed apart from the C terminus. These findings corroborate the small angle x-ray scattering structural model and are instructive for development of high throughput screens that target specific domains and their functions to identify HIV/AIDS therapeutics.


Asunto(s)
Citidina Desaminasa/metabolismo , Proteína p24 del Núcleo del VIH/metabolismo , VIH-1/metabolismo , Modelos Moleculares , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo , Desaminasa APOBEC-3G , Línea Celular , Citidina Desaminasa/química , Citidina Desaminasa/genética , ADN de Cadena Simple/química , ADN de Cadena Simple/genética , ADN de Cadena Simple/metabolismo , ADN Viral/química , ADN Viral/genética , ADN Viral/metabolismo , Dimerización , Proteína p24 del Núcleo del VIH/química , Proteína p24 del Núcleo del VIH/genética , VIH-1/química , VIH-1/genética , Humanos , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína/fisiología , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/química , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética
18.
J Biol Chem ; 283(12): 7320-7, 2008 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-18165230

RESUMEN

Human APOBEC3G (hA3G) is a host factor that defends against HIV-1 as well as other exogenous retroviruses and endogenous retroelements. To this end, hA3G is restricted to the cytoplasm of T lymphocytes where it interacts with viral RNA and proteins to assemble with viral particles causing a post-entry block during reverse transcription. hA3G also exhibits a mechanism to inhibit the reverse transcription of retroelements by RNA binding and sequestration into mRNA processing centers in the cytoplasm. We have determined that the molecular basis for this specialized property of hA3G is a novel cytoplasmic retention signal (CRS) that is necessary and sufficient to restrict wild-type hA3G and chimeric constructs to the cytoplasm. The CRS resides within amino acids 113-128 and is embedded within a basic flanking sequence and does not require RNA binding to retain hA3G in the cytoplasm. Paralogs of hA3G that have nuclear or cytoplasmic distributions differ from hA3G within the region encompassing the CRS motif with respect to charge and amino acid composition. We propose that the CRS enables hA3G to interact with cytoplasmic factors, and thereby enables hA3G to serve in host cell defense by restricting an antiviral sentinel to the cytoplasm. The CRS lies in a region involved in both Gag and Vif interactions; therefore, identification of this motif has important implications for the design of therapeutics that target HIV-1 while maintaining antiviral and cellular functions.


Asunto(s)
Citidina Desaminasa/metabolismo , Citoplasma/metabolismo , VIH-1/metabolismo , Señales de Clasificación de Proteína/fisiología , Procesamiento Postranscripcional del ARN/fisiología , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Desaminasa APOBEC-3G , Secuencias de Aminoácidos/fisiología , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citidina Desaminasa/genética , Citoplasma/genética , VIH-1/genética , Humanos , Transporte de Proteínas/fisiología , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Retroelementos/fisiología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo
19.
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
20.
Biochem Biophys Res Commun ; 350(1): 214-9, 2006 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-16999936

RESUMEN

Human APOBEC3G (hA3G) is a member of the APOBEC-1 related protein (ARP) family of cytidine deaminases. hA3G functions as a natural defense against endogenous retrotransposons and a multitude of retroviruses, most notably human immunodeficiency virus type 1 (HIV-1). Nothing is known about the cellular function of hA3G, however, upon HIV-1 infection hA3G functions as an antiviral factor by mutating viral single-stranded DNA during reverse transcription. Whereas homologous deaminases such as APOBEC-1 and AID act on RNA and DNA, respectively, in the cell nucleus, hA3G mutagenic activity appears to be restricted to the cytoplasm. We demonstrate that hA3G is not a nucleo-cytoplasmic shuttling protein like APOBEC-1 and AID, but is strongly retained in the cytoplasm through a mechanism that involves both the N and C-terminal regions of the protein.


Asunto(s)
Núcleo Celular/metabolismo , Citidina Desaminasa/metabolismo , Citoplasma/metabolismo , Desaminasas APOBEC-1 , Secuencia de Aminoácidos , Citidina Desaminasa/química , Citidina Desaminasa/genética , Células HeLa , Humanos , Leucina/genética , Leucina/metabolismo , Datos de Secuencia Molecular , Transporte de Proteínas , Alineación de Secuencia , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...