Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nature ; 615(7953): 728-733, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36754086

RESUMEN

The APOBEC3 (A3) proteins are host antiviral cellular proteins that hypermutate the viral genome of diverse viral families. In retroviruses, this process requires A3 packaging into viral particles1-4. The lentiviruses encode a protein, Vif, that antagonizes A3 family members by targeting them for degradation. Diversification of A3 allows host escape from Vif whereas adaptations in Vif enable cross-species transmission of primate lentiviruses. How this 'molecular arms race' plays out at the structural level is unknown. Here, we report the cryogenic electron microscopy structure of human APOBEC3G (A3G) bound to HIV-1 Vif, and the hijacked cellular proteins that promote ubiquitin-mediated proteolysis. A small surface explains the molecular arms race, including a cross-species transmission event that led to the birth of HIV-1. Unexpectedly, we find that RNA is a molecular glue for the Vif-A3G interaction, enabling Vif to repress A3G by ubiquitin-dependent and -independent mechanisms. Our results suggest a model in which Vif antagonizes A3G by intercepting it in its most dangerous form for the virus-when bound to RNA and on the pathway to packaging-to prevent viral restriction. By engaging essential surfaces required for restriction, Vif exploits a vulnerability in A3G, suggesting a general mechanism by which RNA binding helps to position key residues necessary for viral antagonism of a host antiviral gene.


Asunto(s)
Desaminasa APOBEC-3G , VIH-1 , Proteolisis , Productos del Gen vif del Virus de la Inmunodeficiencia Humana , Animales , Humanos , Desaminasa APOBEC-3G/antagonistas & inhibidores , Desaminasa APOBEC-3G/química , Desaminasa APOBEC-3G/metabolismo , Desaminasa APOBEC-3G/ultraestructura , VIH-1/metabolismo , VIH-1/patogenicidad , ARN/química , ARN/metabolismo , Ubiquitina/metabolismo , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/química , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/ultraestructura , Microscopía por Crioelectrón , Empaquetamiento del Genoma Viral , Primates/virología
2.
Int J Mol Sci ; 23(9)2022 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-35563460

RESUMEN

The radiosensitization of tumor cells is one of the promising approaches for enhancing radiation damage to cancer cells and limiting radiation effects on normal tissue. In this study, we performed a comprehensive screening of radiosensitization targets in human lung cancer cell line A549 using an shRNA library and identified apolipoprotein B mRNA editing enzyme catalytic subunit 3G (APOBEC3G: A3G) as a candidate target. APOBEC3G is an innate restriction factor that inhibits HIV-1 infection as a cytidine deaminase. APOBEC3G knockdown with siRNA showed an increased radiosensitivity in several cancer cell lines, including pancreatic cancer MIAPaCa2 cells and lung cancer A549 cells. Cell cycle analysis revealed that APOBEC3G knockdown increased S-phase arrest in MIAPaCa2 and G2/M arrest in A549 cells after γ-irradiation. DNA double-strand break marker γH2AX level was increased in APOBEC3G-knocked-down MIAPaCa2 cells after γ-irradiation. Using a xenograft model of A549 in mice, enhanced radiosensitivity by a combination of X-ray irradiation and APOBEC3G knockdown was observed. These results suggest that the functional inhibition of APOBEC3G sensitizes cancer cells to radiation by attenuating the activation of the DNA repair pathway, suggesting that APOBEC3G could be useful as a target for the radiosensitization of cancer therapy.


Asunto(s)
Desaminasa APOBEC-3G , Rayos gamma , Tolerancia a Radiación , Desaminasa APOBEC-3G/antagonistas & inhibidores , Desaminasa APOBEC-3G/farmacología , Animales , Apoptosis , Línea Celular Tumoral , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular , Rayos gamma/uso terapéutico , Humanos , Neoplasias Pulmonares/radioterapia , Ratones , Tolerancia a Radiación/genética , Tolerancia a Radiación/fisiología
3.
J Virol ; 96(4): e0207121, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-34908448

RESUMEN

APOBEC3G (A3G) is a host-encoded cytidine deaminase that potently restricts retroviruses such as HIV-1 and depends on its ability to package into virions. As a consequence of this, HIV-1 protein Vif has evolved to antagonize human A3G by targeting it for ubiquitination and subsequent degradation. There is an ancient arms race between Vif and A3G highlighted by amino acids 128 and 130 in A3G that have evolved under positive selection due to Vif-mediated selective pressure in Old World primates. Nonetheless, not all possible amino acid combinations at these sites have been sampled by nature, and the evolutionary potential of species to resist Vif antagonism is not clear. To explore the evolutionary space of positively selected sites in the Vif-binding region of A3G, we designed a combinatorial mutagenesis screen to introduce all 20 amino acids at sites 128 and 130. Our screen uncovered mutants of A3G with several interesting phenotypes, including loss of antiviral activity and resistance of Vif antagonism. However, HIV-1 Vif exhibited remarkable flexibility in antagonizing A3G 128 and 130 mutants, which significantly reduces viable Vif resistance strategies for hominid primates. Importantly, we find that broadened Vif specificity was conferred through loop 5 adaptations that were required for cross-species adaptation from Old World monkey A3G to hominid A3G. Our evidence suggests that Vif adaptation to novel A3G interfaces during cross-species transmission may train Vif toward broadened specificity that can further facilitate cross-species transmissions and raise the barrier to host resistance. IMPORTANCE APOBEC3G (A3G) is an antiviral protein that potently restricts retroviruses like HIV. In turn, the HIV-1 protein Vif has evolved to antagonize A3G through degradation. Two rapidly evolving sites in A3G confer resistance to unadapted Vif and act as a barrier to cross-species transmission of retroviruses. We recently identified a single amino acid mutation in a simian immunodeficiency virus (SIV) Vif that contributed to the cross-species origins of SIV infecting chimpanzee and, ultimately, the HIV-1 pandemic. This mutation broadened specificity of this Vif to both antagonize the A3G of its host while simultaneously overcoming the A3G barrier in the great apes. In this work, we explore the evolutionary space of human A3G at these rapidly evolving sites to understand if the broadened Vif specificity gained during cross-species transmission confers an advantage to HIV-1 Vif in its host-virus arms race with A3G.


Asunto(s)
Desaminasa APOBEC-3G/antagonistas & inhibidores , VIH-1/fisiología , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Desaminasa APOBEC-3G/genética , Adaptación Fisiológica/genética , Aminoácidos , Animales , Infecciones por VIH/transmisión , Infecciones por VIH/virología , VIH-1/genética , Interacciones Microbiota-Huesped , Humanos , Mutación , Primates , Virus de la Inmunodeficiencia de los Simios/genética , Zoonosis Virales/transmisión , Zoonosis Virales/virología , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética
4.
Viruses ; 13(4)2021 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-33916704

RESUMEN

The ubiquitin-proteasome system plays an important role in the cell under normal physiological conditions but also during viral infections. Indeed, many auxiliary proteins from the (HIV-1) divert this system to its own advantage, notably to induce the degradation of cellular restriction factors. For instance, the HIV-1 viral infectivity factor (Vif) has been shown to specifically counteract several cellular deaminases belonging to the apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like (APOBEC3 or A3) family (A3A to A3H) by recruiting an E3-ubiquitin ligase complex and inducing their polyubiquitination and degradation through the proteasome. Although this pathway has been extensively characterized so far, Vif has also been shown to impede A3s through degradation-independent processes, but research on this matter remains limited. In this review, we describe our current knowledge regarding the degradation-independent inhibition of A3s, and A3G in particular, by the HIV-1 Vif protein, the molecular mechanisms involved, and highlight important properties of this small viral protein.


Asunto(s)
Desaminasa APOBEC-3G/antagonistas & inhibidores , VIH-1/genética , Interacciones Microbiota-Huesped , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , VIH-1/química , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Ubiquitina/metabolismo , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo
5.
Chembiochem ; 21(7): 1028-1035, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31633265

RESUMEN

The APOBEC3 (APOBEC3A-H) enzyme family is part of the human innate immune system that restricts pathogens by scrambling pathogenic single-stranded (ss) DNA by deamination of cytosines to produce uracil residues. However, APOBEC3-mediated mutagenesis of viral and cancer DNA promotes its evolution, thus enabling disease progression and the development of drug resistance. Therefore, APOBEC3 inhibition offers a new strategy to complement existing antiviral and anticancer therapies by making such therapies effective for longer periods of time, thereby preventing the emergence of drug resistance. Here, we have synthesised 2'-deoxynucleoside forms of several known inhibitors of cytidine deaminase (CDA), incorporated them into oligodeoxynucleotides (oligos) in place of 2'-deoxycytidine in the preferred substrates of APOBEC3A, APOBEC3B, and APOBEC3G, and evaluated their inhibitory potential against these enzymes. An oligo containing a 5-fluoro-2'-deoxyzebularine (5FdZ) motif exhibited an inhibition constant against APOBEC3B 3.5 times better than that of the comparable 2'-deoxyzebularine-containing (dZ-containing) oligo. A similar inhibition trend was observed for wild-type APOBEC3A. In contrast, use of the 5FdZ motif in an oligo designed for APOBEC3G inhibition resulted in an inhibitor that was less potent than the dZ-containing oligo both in the case of APOBEC3GCTD and in that of full-length wild-type APOBEC3G.


Asunto(s)
Desaminasa APOBEC-3G/metabolismo , Citidina/análogos & derivados , ADN de Cadena Simple/química , Flúor/química , Desaminasa APOBEC-3G/antagonistas & inhibidores , Desaminasa APOBEC-3G/genética , Secuencia de Bases , Citidina/química , ADN de Cadena Simple/metabolismo , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Mutagénesis , Resonancia Magnética Nuclear Biomolecular , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/metabolismo , Compuestos Organofosforados/química
6.
Biochemistry ; 58(5): 391-400, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30418757

RESUMEN

APOBEC3 enzymes form part of the innate immune system by deaminating cytosine to uracil in single-stranded DNA (ssDNA) and thereby preventing the spread of pathogenic genetic information. However, APOBEC mutagenesis is also exploited by viruses and cancer cells to increase rates of evolution, escape adaptive immune responses, and resist drugs. This raises the possibility of APOBEC3 inhibition as a strategy for augmenting existing antiviral and anticancer therapies. Here we show that, upon incorporation into short ssDNAs, the cytidine nucleoside analogue 2'-deoxyzebularine (dZ) becomes capable of inhibiting the catalytic activity of selected APOBEC variants derived from APOBEC3A, APOBEC3B, and APOBEC3G, supporting a mechanism in which ssDNA delivers dZ to the active site. Multiple experimental approaches, including isothermal titration calorimetry, fluorescence polarization, protein thermal shift, and nuclear magnetic resonance spectroscopy assays, demonstrate nanomolar dissociation constants and low micromolar inhibition constants. These dZ-containing ssDNAs constitute the first substrate-like APOBEC3 inhibitors and, together, comprise a platform for developing nucleic acid-based inhibitors with cellular activity.


Asunto(s)
Desaminasa APOBEC-3G/antagonistas & inhibidores , Citidina Desaminasa/antagonistas & inhibidores , Citidina/análogos & derivados , ADN de Cadena Simple/farmacología , Inhibidores Enzimáticos/farmacología , Proteínas/antagonistas & inhibidores , Desaminasa APOBEC-3G/metabolismo , Citidina/química , Citidina/farmacología , Citidina Desaminasa/metabolismo , ADN de Cadena Simple/química , Inhibidores Enzimáticos/química , Humanos , Antígenos de Histocompatibilidad Menor/metabolismo , Proteínas/metabolismo
7.
Virology ; 504: 1-11, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28131088

RESUMEN

APOBEC3G (A3G) is a cytidine deaminase with potent antiviral activity that is antagonized by Vif. A3G is expressed in a cell type-specific manner and some semi-permissive cells, including A3.01, express A3G but fail to block replication of Vif-null HIV-1. Here we explored the semi-permissive nature of A3.01 cells and found it to be defined exclusively by the levels of A3G. Indeed, minor changes in A3G levels rendered A3.01 cells either fully permissive or non-permissive for Vif-null HIV-1. Our data indicate that A3.01 cells express sub-lethal levels of catalytically active A3G that affects Vif-null HIV-1 at the proviral level but does not completely block virus replication due to purifying selection. Attempts to use the selective pressure exerted by such sub-lethal levels of A3G to select for APOBEC-resistant Vif-null virus capable of replicating in H9 cells failed despite passaging virus for five months, demonstrating that Vif is a critical viral accessory protein.


Asunto(s)
Desaminasa APOBEC-3G/genética , Linfocitos T CD4-Positivos/virología , Provirus/crecimiento & desarrollo , Replicación Viral/genética , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo , Desaminasa APOBEC-3G/antagonistas & inhibidores , Desaminasa APOBEC-3G/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Preescolar , Femenino , Células HEK293 , Infecciones por VIH/virología , VIH-1/genética , Células HeLa , Humanos , Células Jurkat , Provirus/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Viral/genética , Análisis de Secuencia de ARN , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética
8.
J Mol Biol ; 429(1): 88-96, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-27887868

RESUMEN

The human APOBEC3G (A3G) enzyme restricts HIV-1 in the absence of the viral accessory protein viral infectivity factor (Vif) by deaminating viral cDNA cytosines to uracils. These uracil lesions base-pair with adenines during the completion of reverse transcription and result in A3G signature G-to-A mutations in the viral genome. Vif protects HIV-1 from A3G-mediated restriction by forming an E3-ubiquitin ligase complex to polyubiquitinate A3G and trigger its degradation. Prior studies indicated that Vif may also directly block the enzymatic activity of A3G and, provocatively, that Vif-derived peptides, Vif 25-39 and Vif 105-119, are similarly inhibitory. Here, we show that Vif 25-39 does not inhibit A3G enzymatic activity and that the inhibitory effect of Vif 105-119 and that of a shorter derivative Vif 107-115, although recapitulated, are non-specific. We also elaborate a simple method for assaying DNA cytosine deaminase activity that eliminates potential polymerase chain reaction-induced biases. Our results show that these Vif-derived peptides are unlikely to be useful as tools to study A3G function or as leads for the development of future therapeutics.


Asunto(s)
Desaminasa APOBEC-3G/antagonistas & inhibidores , Inhibidores Enzimáticos/metabolismo , Péptidos/metabolismo , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo , Péptidos/genética , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética
9.
J Mol Biol ; 428(17): 3514-28, 2016 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-27289067

RESUMEN

The APOBEC3 locus consists of seven genes (A3A-A3C, A3DE, A3F-A3H) that encode DNA cytidine deaminases. These enzymes deaminate single-stranded DNA, the result being DNA peppered with CG →TA mutations preferentially in the context of 5'TpC with the exception of APOBEC3G (A3G), which prefers 5'CpC dinucleotides. Hepatitis B virus (HBV) DNA is vulnerable to genetic editing by APOBEC3 cytidine deaminases, A3G being a major restriction factor. APOBEC3DE (A3DE) stands out in that it is catalytically inactive due to a fixed Tyr320Cys substitution in the C-terminal domain. As A3DE is closely related to A3F and A3G, which can form homo- and heterodimers and multimers, the impact of A3DE on HBV replication via modulation of other APOBEC3 restriction factors was investigated. A3DE binds to itself, A3F, and A3G and antagonizes A3F and, to a lesser extent, A3G restriction of HBV replication. A3DE suppresses A3F and A3G from HBV particles, leading to enhanced HBV replication. Ironically, while being part of a cluster of innate restriction factors, the A3DE phenotype is proviral. As the gorilla genome encodes the same Tyr320Cys substitution, this proviral phenotype seems to have been selected for.


Asunto(s)
Desaminasa APOBEC-3G/antagonistas & inhibidores , Citidina Desaminasa/metabolismo , Citosina Desaminasa/antagonistas & inhibidores , Virus de la Hepatitis B/inmunología , Virus de la Hepatitis B/fisiología , Replicación Viral , Desaminasa APOBEC-3G/metabolismo , Animales , Línea Celular , Citosina Desaminasa/metabolismo , Gorilla gorilla , Humanos , Unión Proteica
10.
AIDS ; 30(11): 1723-9, 2016 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-27064995

RESUMEN

OBJECTIVE: The human cytidine deaminase APOBEC3G (A3G) potently restricts HIV-1 but the virus, in turn, expresses a Vif protein which degrades A3G. A natural A3G-H186R variant, common in African populations, has been associated with a more rapid AIDS disease progression, but the underlying mechanism remains unknown. We hypothesized that differences in HIV-1 Vif activity towards A3G wild type and A3G-H186R contribute to the distinct clinical AIDS manifestation. METHODS: Vif variants were cloned from plasma samples of 26 South African HIV-1 subtype C infected patients, which either express wild type A3G or A3G-H186R. The Vif alleles were assessed for their ability to counteract A3G variants using western blot and single-cycle infectivity assays. RESULTS: We obtained a total of 392 Vif sequences which displayed an amino acid sequence difference of 6.2-19.2% between patients. The intrapatient Vif diversities from patient groups A3G, A3G and A3G were similar. Vif variants obtained from patients expressing A3G and A3G were capable of counteracting both A3G variants with similar efficiency. However, the antiviral activity of A3G-H186R was significantly reduced in both the presence and absence of Vif, indicating that the A3G-H186R variant intrinsically exerts less antiviral activity. CONCLUSION: A3G wild type and A3G-H186R are equally susceptible to counteraction by Vif, regardless of whether the Vif variant was obtained from A3G and A3G patients. However, the A3G-H186R variant intrinsically displayed lower antiviral activity, which could explain the higher plasma viral loads and accelerated disease progression reported for patients expressing A3G.


Asunto(s)
Desaminasa APOBEC-3G/antagonistas & inhibidores , Desaminasa APOBEC-3G/genética , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Haplotipos , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo , Femenino , Humanos , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA