Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
1.
J Phys Chem B ; 128(13): 3102-3112, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38516924

RESUMO

The yeast cytosine deaminase (yCD) enzyme/5-fluorocytosine prodrug system is a promising candidate for targeted chemotherapeutics. After conversion of the prodrug into the toxic chemotherapeutic drug, 5-fluorouracil (5-FU), the slow product release from the enzyme limits the overall catalytic efficiency of the enzyme/prodrug system. Here, we present a computational study of the product release of the anticancer drug, 5-FU, from yCD using metadynamics. We present a comparison of the 5-FU drug to the natural enzyme product, uracil. We use volume-based metadynamics to compute the free energy landscape for product release and show a modest binding affinity for the product to the enzyme, consistent with experiments. Next, we use infrequent metadynamics to estimate the unbiased release rate from Kramers time-dependent rate theory and find a favorable comparison to experiment with a slower rate of product release for the 5-FU system. Our work demonstrates how adaptive sampling methods can be used to study the protein-ligand unbinding process for engineering enzyme/prodrug systems and gives insights into the molecular mechanism of product release for the yCD/5-FU system.


Assuntos
Antineoplásicos , Pró-Fármacos , Saccharomyces cerevisiae , Citosina Desaminase/química , Citosina Desaminase/metabolismo , Fluoruracila/metabolismo , Flucitosina/química , Flucitosina/metabolismo , Pró-Fármacos/química
2.
J Infect Dis ; 228(10): 1421-1429, 2023 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-37224627

RESUMO

BACKGROUND: On May 6, 2022, a powerful outbreak of monkeypox virus (MPXV) had been reported outside of Africa, with many continuing new cases being reported around the world. Analysis of mutations among the 2 different lineages present in the 2021 and 2022 outbreaks revealed the presence of G->A mutations occurring in the 5'GpA context, indicative of APOBEC3 cytidine deaminase activity. METHODS: By using a sensitive polymerase chain reaction (differential DNA denaturation PCR) method allowing differential amplification of AT-rich DNA, we analyzed the level of APOBEC3-induced MPXV editing in infected cells and in patients. RESULTS: We demonstrate that G->A hypermutated MPXV genomes can be recovered experimentally from APOBEC3 transfection followed by MPXV infection. Here, among the 7 human APOBEC3 cytidine deaminases (A3A-A3C, A3DE, A3F-A3H), only APOBEC3F was capable of extensively deaminating cytidine residues in MPXV genomes. Hyperedited genomes were also recovered in ∼42% of analyzed patients. Moreover, we demonstrate that substantial repair of these mutations occurs. Upon selection, corrected G->A mutations escaping drift loss contribute to the MPXV evolution observed in the current epidemic. CONCLUSIONS: Stochastic or transient overexpression of the APOBEC3F gene exposes the MPXV genome to a broad spectrum of mutations that may be modeling the mutational landscape after multiple cycles of viral replication.


Assuntos
Citidina Desaminase , Monkeypox virus , Humanos , Monkeypox virus/genética , Citidina Desaminase/genética , Mutação , Surtos de Doenças , Citidina , Citosina Desaminase/química , Citosina Desaminase/genética
3.
Anal Chim Acta ; 1281: 341895, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-38783732

RESUMO

APOBEC3A (A3A) is a cytidine deaminase with critical roles in molecular diagnostics. Herein, we demonstrate the enzymatic DNA repairing amplification-powered construction of an Au nanoparticle-based nanosensor for single-molecule monitoring of A3A activity in cancer cells. Target A3A can convert cytosine (C) in substrate probe to uracil (U), and then the template binds with substrate probe to form a dsDNA containing U/A base pairs. Uracil DNA glycosylase (UDG) excises the U base to produce an apurinic/apyrimidinic (AP) site that can be cleaved by apurinic/apyrimidic endonuclease 1 (APE1) to obtain the substrate fragment with 3'-OH end. Subsequently, the substrate fragment initiates cyclic enzymatic repairing amplification (ERA), releasing trigger-1 and trigger-2. The resultant trigger-1 can act as the primer to induce multiple cycles of cyclic ERA, producing numerous trigger-1 and trigger-2. The hybridization of trigger-2 with signal probe forms the dsDNA duplexes with an AP site, inducing the cyclic cleavage of signal probes by APE1 to release abundant Cy5 molecules from the AuNPs. Released Cy5 molecules can be easily quantified by single-molecule imaging. This nanosensor allows for specific and sensitive detection of A3A activity with a detection limit of 0.855 aM, and it can further measure kinetic parameters, screen inhibitors, and quantify endogenous A3A activity at the single-cell level, with prospect application in disease diagnostics and therapy.


Assuntos
Ouro , Nanopartículas Metálicas , Ouro/química , Nanopartículas Metálicas/química , Humanos , Técnicas Biossensoriais/métodos , Reparo do DNA , Técnicas de Amplificação de Ácido Nucleico , Citosina Desaminase/metabolismo , Citosina Desaminase/química , DNA/química , Imagem Individual de Molécula/métodos , DNA Liase (Sítios Apurínicos ou Apirimidínicos)
4.
J Virol ; 95(12)2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-33789992

RESUMO

APOBEC3 proteins play pivotal roles in defenses against retroviruses, including HIV-1, as well as retrotransposons. Presumably due to the evolutionary arms race between the hosts and retroelements, APOBEC3 genes have rapidly evolved in primate lineages through sequence diversification, gene amplification and loss, and gene fusion. Consequently, modern primates possess a unique set or "repertoire" of APOBEC3 genes. The APOBEC3 gene repertoire of humans has been well investigated. There are three types of catalytic domains (Z domain; A3Z1, A3Z2, and A3Z3), 11 Z domains, and 7 independent genes, including 4 genes encoding double Z domains. However, the APOBEC3 gene repertoires of nonhuman primates remain largely unclear. Here, we characterize APOBEC3 gene repertoires among primates and investigated the evolutionary scenario of primate APOBEC3 genes using phylogenetic and comparative genomics approaches. In the 21 primate species investigated, we identified 145 APOBEC3 genes, including 69 double-domain type APOBEC3 genes. We further estimated the ages of the respective APOBEC3 genes and revealed that APOBEC3B, APOBEC3D, and APOBEC3F are the youngest in humans and were generated in the common ancestor of Catarrhini. Notably, invasion of the LINE1 retrotransposon peaked during the same period as the generation of these youngest APOBEC3 genes, implying that LINE1 invasion was one of the driving forces of the generation of these genes. Moreover, we found evidence suggesting that sequence diversification by gene conversions among APOBEC3 paralogs occurred in multiple primate lineages. Together, our analyses reveal the hidden diversity and the complicated evolutionary scenario of APOBEC3 genes in primates.IMPORTANCE In terms of virus-host interactions and coevolution, the APOBEC3 gene family is one of the most important subjects in the field of retrovirology. APOBEC3 genes are composed of a repertoire of subclasses based on sequence similarity, and a paper by LaRue et al. provides the standard guideline for the nomenclature and genomic architecture of APOBEC3 genes. However, it has been more than 10 years since this publication, and new information, including RefSeq, which we used in this study, is accumulating. Based on accumulating knowledge, APOBEC3 genes, particularly those of primates, should be refined and reannotated. This study updates knowledge of primate APOBEC3 genes and their genomic architectures. We further inferred the evolutionary scenario of primate APOBEC3 genes and the potential driving forces of APOBEC3 gene evolution. This study will be a landmark for the elucidation of the multiple aspects of APOBEC3 family genes in the future.


Assuntos
Desaminases APOBEC/genética , Evolução Molecular , Primatas/genética , Desaminases APOBEC/química , Animais , Domínio Catalítico , Citidina Desaminase/química , Citidina Desaminase/genética , Citosina Desaminase/química , Citosina Desaminase/genética , Conversão Gênica , Humanos , Elementos Nucleotídeos Longos e Dispersos , Antígenos de Histocompatibilidade Menor/química , Antígenos de Histocompatibilidade Menor/genética , Filogenia
5.
Int J Mol Sci ; 22(4)2021 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-33672989

RESUMO

Epidermal growth factor receptor (EGFR) specific therapeutics is of great importance in cancer treatment. Fcy-hEGF fusion protein, composed of yeast cytosine deaminase (Fcy) and human EGF (hEGF), is capable of binding to EGFR and enzymatically convert 5-fluorocytosine (5-FC) to 1000-fold toxic 5-fluorocuracil (5-FU), thereby inhibiting the growth of EGFR-expressing tumor cells. To develop EGFR-specific therapy, 188Re-liposome-Fcy-hEGF was constructed by insertion of Fcy-hEGF fusion protein onto the surface of liposomes encapsulating of 188Re. Western blotting, MALDI-TOF, column size exclusion and flow cytometry were used to confirm the conjugation and bio-activity of 188Re-liposome-Fcy-hEGF. Cell lines with EGFR expression were subjected to treat with 188Re-liposome-Fcy-hEGF/5-FC in the presence of 5-FC. The 188Re-liposome-Fcy-hEGF/5-FC revealed a better cytotoxic effect for cancer cells than the treatment of liposome-Fcy-hEGF/5-FC or 188Re-liposome-Fcy-hEGF alone. The therapeutics has radio- and chemo-toxicity simultaneously and specifically target to EGFR-expression tumor cells, thereby achieving synergistic anticancer activity.


Assuntos
Citosina Desaminase/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Fluoruracila/farmacologia , Neoplasias/metabolismo , Compostos Radiofarmacêuticos/farmacologia , Antimetabólitos Antineoplásicos/metabolismo , Antimetabólitos Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Citosina Desaminase/química , Fator de Crescimento Epidérmico/química , Flucitosina/metabolismo , Fluoruracila/metabolismo , Humanos , Lipossomos/química , Células MCF-7 , Neoplasias/patologia , Ligação Proteica , Radioisótopos/química , Compostos Radiofarmacêuticos/química , Rênio/química
6.
Nucleic Acids Res ; 48(3): 1353-1371, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31943071

RESUMO

The human apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like 3 (APOBEC3, A3) family member proteins can deaminate cytosines in single-strand (ss) DNA, which restricts human immunodeficiency virus type 1 (HIV-1), retrotransposons, and other viruses such as hepatitis B virus, but can cause a mutator phenotype in many cancers. While structural information exists for several A3 proteins, the precise details regarding deamination target selection are not fully understood. Here, we report the first parallel, comparative analysis of site selection of A3 deamination using six of the seven purified A3 member enzymes, oligonucleotides having 5'TC3' or 5'CT3' dinucleotide target sites, and different flanking bases within diverse DNA secondary structures. A3A, A3F and A3H were observed to have strong preferences toward the TC target flanked by A or T, while all examined A3 proteins did not show a preference for a TC target flanked by a G. We observed that the TC target was strongly preferred in ssDNA regions rather than dsDNA, loop or bulge regions, with flanking bases influencing the degree of preference. CT was also shown to be a potential deamination target. Taken together, our observations provide new insights into A3 enzyme target site selection and how A3 mutagenesis impacts mutation rates.


Assuntos
Citidina Desaminase/genética , DNA de Cadeia Simples/genética , Proteínas de Ligação a DNA/genética , Desaminação/genética , Desaminases APOBEC , Sítios de Ligação/genética , Linhagem Celular , Citidina Desaminase/química , Citosina Desaminase/química , Citosina Desaminase/genética , DNA de Cadeia Simples/química , Proteínas de Ligação a DNA/química , HIV-1/genética , HIV-1/patogenicidade , Vírus da Hepatite B/genética , Humanos , Mutagênese/genética , Conformação de Ácido Nucleico , Estrutura Secundária de Proteína , Retroelementos/genética
7.
Biochim Biophys Acta Gen Subj ; 1864(2): 129346, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-30986508

RESUMO

BACKGROUND: APOBEC3F (A3F), a member of the human APOBEC3 (A3) family of cytidine deaminases, acts as an anti-HIV-1 factor by deaminating deoxycytidine in the complementary DNA of the viral genome. A full understanding of the deamination behavior of A3F awaits further investigation. METHODS: The real-time NMR method and uracil-DNA glycosylase assay were used to track the activities of the C-terminal domain (CTD) of A3F at different concentrations of A3F-CTD and ssDNA. The steady-state fluorescence anisotropy measurement was used to examine the binding between A3F-CTD and ssDNA with different lengths. The use of the A3F-CTD N214H mutant, having higher activity than the wild-type, facilitated the tracking of the reactions. RESULTS: A3F-CTD was found to efficiently deaminate the target deoxycytidine in long ssDNA in lower ssDNA concentration conditions ([A3F-CTD] ≫ [ssDNA]), while the target deoxycytidine in short ssDNA is deaminated efficiently in higher ssDNA concentration conditions ([A3F-CTD] ≪ [ssDNA]). This property is quite different from that of the previously studied A3 family member, A3B; the concentrations of the proteins and ssDNA had no effect. CONCLUSIONS: The concentrations of A3F-CTD and ssDNA substrates affect the ssDNA-length-dependence of deamination rate of the A3F-CTD. This unique property of A3F is rationally interpreted on the basis of its binding characteristics with ssDNA. GENERAL SIGNIFICANCE: The discovery of the unique property of A3F regarding the deamination rate deepens the understanding of its counteraction against HIV-1. Our strategy is applicable to investigate the other aspects of the A3 activities, such as those involved in the cancer development.


Assuntos
Citosina Desaminase/química , DNA de Cadeia Simples/química , Citidina Desaminase/química , Desaminação , Genoma Viral , HIV-1/genética , Humanos , Cinética , Espectroscopia de Ressonância Magnética , Conformação Molecular , Mutação , Oligonucleotídeos/química , Ligação Proteica
8.
Nat Struct Mol Biol ; 26(12): 1176-1183, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31792451

RESUMO

HIV-1 virion infectivity factor (Vif) promotes degradation of the antiviral APOBEC3 (A3) proteins through the host ubiquitin-proteasome pathway to enable viral immune evasion. Disrupting Vif-A3 interactions to reinstate the A3-catalyzed suppression of human immunodeficiency virus type 1 (HIV-1) replication is a potential approach for antiviral therapeutics. However, the molecular mechanisms by which Vif recognizes A3 proteins remain elusive. Here we report a cryo-EM structure of the Vif-targeted C-terminal domain of human A3F in complex with HIV-1 Vif and the cellular cofactor core-binding factor beta (CBFß) at 3.9-Å resolution. The structure shows that Vif and CBFß form a platform to recruit A3F, revealing a direct A3F-recruiting role of CBFß beyond Vif stabilization, and captures multiple independent A3F-Vif interfaces. Together with our biochemical and cellular studies, our structural findings establish the molecular determinants that are critical for Vif-mediated neutralization of A3F and provide a comprehensive framework of how HIV-1 Vif hijacks the host protein degradation machinery to counteract viral restriction by A3F.


Assuntos
Citosina Desaminase/química , HIV-1/química , Produtos do Gene vif do Vírus da Imunodeficiência Humana/química , Subunidade beta de Fator de Ligação ao Core/química , Microscopia Crioeletrônica , Citosina Desaminase/antagonistas & inibidores , Citosina Desaminase/ultraestrutura , Humanos , Evasão da Resposta Imune , Modelos Moleculares , Conformação Proteica , Domínios Proteicos , Mapeamento de Interação de Proteínas , Proteólise , Relação Estrutura-Atividade , Produtos do Gene vif do Vírus da Imunodeficiência Humana/farmacologia , Produtos do Gene vif do Vírus da Imunodeficiência Humana/ultraestrutura
9.
Science ; 365(6451): 382-386, 2019 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-31296651

RESUMO

Programmable RNA editing enables reversible recoding of RNA information for research and disease treatment. Previously, we developed a programmable adenosine-to-inosine (A-to-I) RNA editing approach by fusing catalytically inactivate RNA-targeting CRISPR-Cas13 (dCas13) with the adenine deaminase domain of ADAR2. Here, we report a cytidine-to-uridine (C-to-U) RNA editor, referred to as RNA Editing for Specific C-to-U Exchange (RESCUE), by directly evolving ADAR2 into a cytidine deaminase. RESCUE doubles the number of mutations targetable by RNA editing and enables modulation of phosphosignaling-relevant residues. We apply RESCUE to drive ß-catenin activation and cellular growth. Furthermore, RESCUE retains A-to-I editing activity, enabling multiplexed C-to-U and A-to-I editing through the use of tailored guide RNAs.


Assuntos
Adenosina Desaminase/genética , Citidina/genética , Citosina Desaminase/genética , Engenharia de Proteínas/métodos , Edição de RNA , Proteínas de Ligação a RNA/genética , Uridina/genética , Adenosina/genética , Adenosina Desaminase/química , Citosina Desaminase/química , Células HEK293 , Humanos , Inosina/genética , Domínios Proteicos , Proteínas de Ligação a RNA/química , beta Catenina/química , beta Catenina/genética , beta Catenina/metabolismo
10.
Biomacromolecules ; 20(4): 1683-1690, 2019 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-30884222

RESUMO

The simultaneous delivery of multiple therapeutics to a single site has shown promise for cancer targeting and treatment. However, because of the inherent differences in charge and size between drugs and biomolecules, new approaches are required for colocalization of unlike components in one delivery vehicle. In this work, we demonstrate that triblock copolymers containing click nucleic acids (CNAs) can be used to simultaneously load a prodrug enzyme (cytosine deaminase, CodA) and a chemotherapy drug (doxorubicin, DOX) in a single polymer nanoparticle. CNAs are synthetic analogs of DNA comprised of a thiolene backbone and nucleotide bases that can hybridize to complementary strands of DNA. In this study, CodA was appended with complementary DNA sequences and fluorescent dyes to allow its encapsulation in PEG-CNA-PLGA nanoparticles. The DNA-modified CodA was found to retain its enzyme activity for converting prodrug 5-fluorocytosine (5-FC) to active 5-fluorouracil (5-FU) using a modified fluorescent assay. The DNA-conjugated CodA was then loaded into the PEG-CNA-PLGA nanoparticles and tested for cell cytotoxicity in the presence of the 5-FC prodrug. To study the effect of coloading DOX and CodA within a single nanoparticle, cell toxicity assays were run to compare dually loaded nanoparticles with nanoparticles loaded only with either DOX or CodA. We show that the highest level of cell death occurred when both DOX and CodA were simultaneously entrapped and delivered to cells in the presence of 5-FC.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Citosina Desaminase , DNA , Portadores de Fármacos , Enzimas Imobilizadas , Proteínas de Escherichia coli , Nanopartículas , Neoplasias , Poliésteres , Polietilenoglicóis , Pró-Fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Linhagem Celular Tumoral , Citosina Desaminase/química , Citosina Desaminase/farmacologia , DNA/química , DNA/farmacologia , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacologia , Portadores de Fármacos/química , Portadores de Fármacos/uso terapêutico , Enzimas Imobilizadas/química , Enzimas Imobilizadas/farmacologia , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/farmacologia , Flucitosina/química , Flucitosina/farmacocinética , Flucitosina/farmacologia , Humanos , Nanopartículas/química , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Poliésteres/síntese química , Poliésteres/química , Poliésteres/farmacologia , Polietilenoglicóis/síntese química , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Pró-Fármacos/química , Pró-Fármacos/farmacologia
11.
Chembiochem ; 20(15): 1898-1905, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30809902

RESUMO

Over the past few decades, various DNA modification detection methods have been developed; many of the high-resolution methods are based on bisulfite treatment, which leads to DNA degradation, to a degree. Thus, novel bisulfite-free approaches have been developed in recent years and shown to be useful for epigenome analysis in otherwise difficult-to-handle, but important, DNA samples, such as hmC-seal and hmC-CATCH. Herein, an overview of advances in the development of epigenome sequencing methods for these important DNA modifications is provided.


Assuntos
Citosina/metabolismo , Citosina/química , Citosina Desaminase/química , Citosina Desaminase/metabolismo , DNA/química , DNA/genética , DNA/metabolismo , Humanos , Estrutura Molecular
12.
Bioconjug Chem ; 30(2): 432-442, 2019 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-30615416

RESUMO

Proteins are ideal candidates for disease treatment because of their high specificity and potency. Despite this potential, delivery of proteins remains a significant challenge due to the intrinsic size, charge, and stability of proteins. Attempts to overcome these challenges have most commonly relied on direct conjugation of polymers and peptides to proteins via reactive groups on naturally occurring residues. While such approaches have shown some success, they allow limited control of the spacing and number of moieties coupled to proteins, which can hinder bioactivity and delivery capabilities of the therapeutic. Here, we describe a strategy to site-specifically conjugate delivery moieties to therapeutic proteins through unnatural amino acid (UAA) incorporation, in order to explore the effect of epidermal growth factor receptor (EGFR)-targeted ligand valency and spacing on internalization of proteins in EGFR-overexpressing inflammatory breast cancer (IBC) cells. Our results demonstrate the ability to enhance targeted protein delivery by tuning a small number of EGFR ligands per protein and clustering these ligands to promote multivalent ligand-receptor interactions. Furthermore, the tailorability of this simple approach was demonstrated through IBC-targeted cell death via the delivery of yeast cytosine deaminase (yCD), a prodrug converting enzyme.


Assuntos
Aminoácidos/metabolismo , Citosina Desaminase/administração & dosagem , Proteínas Luminescentes/administração & dosagem , Aminoácidos/química , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Química Click , Citosina Desaminase/química , Citosina Desaminase/farmacocinética , Sistemas de Liberação de Medicamentos , Receptores ErbB/metabolismo , Feminino , Humanos , Ligantes , Proteínas Luminescentes/química , Proteínas Luminescentes/farmacocinética , Modelos Moleculares , Ligação Proteica , Leveduras/enzimologia , Proteína Vermelha Fluorescente
13.
Virology ; 527: 21-31, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30448640

RESUMO

The APOBEC3 enzyme family are host restriction factors that induce mutagenesis of HIV-1 proviral genomes through the deamination of cytosine to form uracil in nascent single-stranded (-)DNA. HIV-1 suppresses APOBEC3 activity through the HIV-1 protein Vif that induces APOBEC3 degradation. Here we compared two common polymorphisms of APOBEC3F. We found that although both polymorphisms have HIV-1 restriction activity, APOBEC3F 108 A/231V can restrict HIV-1 ΔVif up to 4-fold more than APOBEC3F 108 S/231I and is partially protected from Vif-mediated degradation. This resulted from higher levels of steady state expression of APOBEC3F 108 A/231 V. Individuals are commonly heterozygous for the APOBEC3F polymorphisms and these polymorphisms formed in cells, independent of RNA, hetero-oligomers between each other and with APOBEC3G. Hetero-oligomerization with APOBEC3F 108 A/231V resulted in partial stabilization of APOBEC3F 108 S/231I and APOBEC3G in the presence of Vif. These data demonstrate functional outcomes of APOBEC3 polymorphisms and hetero-oligomerization that affect HIV-1 restriction.


Assuntos
Citosina Desaminase/genética , Infecções por HIV/genética , HIV-1/genética , Polimorfismo Genético , Replicação Viral , Desaminase APOBEC-3G/química , Desaminase APOBEC-3G/genética , Citosina Desaminase/química , DNA Viral/genética , Células HEK293 , HIV-1/fisiologia , Heterozigoto , Humanos , Mutação , Multimerização Proteica , Estabilidade Proteica , Vírion/metabolismo , Produtos do Gene vif do Vírus da Imunodeficiência Humana/genética , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo
14.
Biomacromolecules ; 19(10): 3945-3957, 2018 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-30160482

RESUMO

The simultaneous expression in Escherichia coli cells of the Qß virus-like particle (VLP) capsid protein and protein "cargo" tagged with a positively charged Rev peptide sequence leads to the spontaneous self-assembly of VLPs with multiple copies of the cargo inside. We report the packaging of four new enzymes with potential applications in medicine and chemical manufacturing. The captured enzymes are active while inside the nanoparticle shell and are protected from environmental conditions that lead to free-enzyme destruction. We also describe genetic modifications to the packaging scheme that shed light on the self-assembly mechanism of this system and allow indirect control over the internal packaging density of cargo. The technology was extended to create, via self-assembly, VLPs that simultaneously display protein ligands on the exterior and contain enzymes within. Inverse relationships were observed between the size of both the packaged and externally displayed protein or domains and nanoparticle yield. These results provide a general method for the rapid creation of robust protein nanoparticles with desired catalytic and targeting functionalities.


Assuntos
Proteínas do Capsídeo/metabolismo , Produtos do Gene rev/metabolismo , Enzimas Multifuncionais/química , Enzimas Multifuncionais/metabolismo , Nanopartículas/metabolismo , RNA Viral/metabolismo , Montagem de Vírus , Aldeído Liases/química , Aldeído Liases/genética , Aldeído Liases/metabolismo , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Catálise , Citosina Desaminase/química , Citosina Desaminase/genética , Citosina Desaminase/metabolismo , Produtos do Gene rev/química , Produtos do Gene rev/genética , Células HeLa , Humanos , Enzimas Multifuncionais/genética , Nanopartículas/química , RNA Viral/química , RNA Viral/genética
15.
Retrovirology ; 15(1): 31, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29636069

RESUMO

BACKGROUND: The apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3 (APOBEC3; A3) gene family appears only in mammalian genomes. Some A3 proteins can be incorporated into progeny virions and inhibit lentiviral replication. In turn, the lentiviral viral infectivity factor (Vif) counteracts the A3-mediated antiviral effect by degrading A3 proteins. Recent investigations have suggested that lentiviral vif genes evolved to combat mammalian APOBEC3 proteins, and have further proposed that the Vif-A3 interaction may help determine the co-evolutionary history of cross-species lentiviral transmission in mammals. RESULTS: Here we address the co-evolutionary relationship between two New World felids, the puma (Puma concolor) and the bobcat (Lynx rufus), and their lentiviruses, which are designated puma lentiviruses (PLVs). We demonstrate that PLV-A Vif counteracts the antiviral action of APOBEC3Z3 (A3Z3) of both puma and bobcat, whereas PLV-B Vif counteracts only puma A3Z3. The species specificity of PLV-B Vif is irrespective of the phylogenic relationships of feline species in the genera Puma, Lynx and Acinonyx. We reveal that the amino acid at position 178 in the puma and bobcat A3Z3 is exposed on the protein surface and determines the sensitivity to PLV-B Vif-mediated degradation. Moreover, although both the puma and bobcat A3Z3 genes are polymorphic, their sensitivity/resistance to PLV Vif-mediated degradation is conserved. CONCLUSIONS: To the best of our knowledge, this is the first study suggesting that the host A3 protein potently controls inter-genus lentiviral transmission. Our findings provide the first evidence suggesting that the co-evolutionary arms race between lentiviruses and mammals has occurred in the New World.


Assuntos
Citosina Desaminase/genética , Interações Hospedeiro-Patógeno/genética , Infecções por Lentivirus/transmissão , Infecções por Lentivirus/virologia , Lentivirus/fisiologia , Animais , Gatos , Citosina Desaminase/química , Citosina Desaminase/metabolismo , Resistência à Doença , Evolução Molecular , Produtos do Gene vif , Vírus da Imunodeficiência Felina/classificação , Vírus da Imunodeficiência Felina/genética , Lentivirus/classificação , Mutação com Perda de Função , Modelos Moleculares , Filogenia , Polimorfismo Genético , Conformação Proteica , Proteólise , Relação Estrutura-Atividade , Treonina/química , Treonina/genética
17.
Nat Struct Mol Biol ; 25(1): 45-52, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29323274

RESUMO

The APOBEC-AID family of cytidine deaminase prefers single-stranded nucleic acids for cytidine-to-uridine deamination. Single-stranded nucleic acids are commonly involved in the DNA repair system for breaks generated by CRISPR-Cas9. Here, we show in human cells that APOBEC3 can trigger cytidine deamination of single-stranded oligodeoxynucleotides, which ultimately results in base substitution mutations in genomic DNA through homology-directed repair (HDR) of Cas9-generated double-strand breaks. In addition, the APOBEC3-catalyzed deamination in genomic single-stranded DNA formed during the repair of Cas9 nickase-generated single-strand breaks in human cells can be further processed to yield mutations mainly involving insertions or deletions (indels). Both APOBEC3-mediated deamination and DNA-repair proteins play important roles in the generation of these indels. Therefore, optimizing conditions for the repair of CRISPR-Cas9-generated DNA breaks, such as using double-stranded donors in HDR or temporarily suppressing endogenous APOBEC3s, can repress these unwanted mutations in genomic DNA.


Assuntos
Sistemas CRISPR-Cas , Citosina Desaminase/química , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Mutação , Desaminases APOBEC , Citidina/química , Citidina Desaminase/genética , DNA de Cadeia Simples , Células HEK293 , Células HeLa , Humanos , Mutação INDEL , Oligonucleotídeos/genética , RNA Interferente Pequeno/metabolismo , Reparo de DNA por Recombinação , Análise de Sequência de DNA
18.
Phys Chem Chem Phys ; 20(5): 3109-3117, 2018 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-28825755

RESUMO

APOBEC3F (A3F), an apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3 (APOBEC3) family protein, catalyzes cytosine-to-uracil conversion in single-stranded (ss) DNA. A3F acts as an inhibitor of retrovirus replication and exhibits antiviral activity against viral infectivity factor (Vif)-deficient human immunodeficiency virus 1 (HIV-1). Previous studies have mostly been focused on the interaction between A3F and Vif, and the studies on A3F's deamination properties are limited. Here, we report comprehensive characterization of the deaminase activity and ssDNA binding of the C-terminal domain (CTD) of A3F. It was shown that the deaminase activity of A3F-CTD is affected by the nucleic acid residues adjacent to the target sequence, TC, and that TTCA/G are the most preferred sequences. A3F-CTD deaminates the target sequence in longer ssDNAs most efficiently. Mutation analysis identified the amino acid residues that are responsible for the deaminase activity and ssDNA binding in the loops surrounding the catalytic center. The functions of these residues were rationally interpreted on the basis of the co-crystal structure of A3A-ssDNA and the known roles of the equivalent amino acid residues found in other A3s. Furthermore, we demonstrated that the deaminase activity of A3F-CTD could be regulated through phosphorylation of a putative site, S216. Finally, A3F-CTD was found to be active in a wide pH range (5.5 to 9.5) with similar activity. Interestingly, the A3F-CTD N214H mutant exhibited a dramatic increase in activity at pH 5.5.


Assuntos
Aminoácidos/metabolismo , Citosina Desaminase/química , Sequência de Aminoácidos , Aminoácidos/química , Sequência de Bases , Sítios de Ligação , Biocatálise , Domínio Catalítico , Cristalografia por Raios X , Citosina Desaminase/genética , Citosina Desaminase/metabolismo , DNA de Cadeia Simples/metabolismo , Desaminação , Polarização de Fluorescência , Humanos , Concentração de Íons de Hidrogênio , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Fosforilação , Ligação Proteica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação
19.
J Am Chem Soc ; 140(1): 14-17, 2018 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-29251923

RESUMO

There is growing interest in designing spatiotemporal control over enzyme activities using noninvasive stimuli such as light. Here, we describe a structure-based, computation-guided predictive method for reversibly controlling enzyme activity using covalently attached photoresponsive azobenzene groups. Applying the method to the therapeutically useful enzyme yeast cytosine deaminase, we obtained a ∼3-fold change in enzyme activity by the photocontrolled modulation of the enzyme's active site lid structure, while fully maintaining thermostability. Multiple cycles of switching, controllable in real time, are possible. The predictiveness of the method is demonstrated by the construction of a variant that does not photoswitch as expected from computational modeling. Our design approach opens new avenues for optically controlling enzyme function. The designed photocontrolled cytosine deaminases may also aid in improving chemotherapy approaches that utilize this enzyme.


Assuntos
Compostos Azo/química , Citosina Desaminase/química , Citosina Desaminase/efeitos da radiação , Processos Fotoquímicos , Compostos Azo/metabolismo , Citosina Desaminase/metabolismo , Modelos Moleculares , Saccharomyces cerevisiae/enzimologia
20.
Viruses ; 9(11)2017 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-29149056

RESUMO

Intrinsic factors of the innate immune system include the apolipoprotein B editing enzyme catalytic polypeptide-like 3 (APOBEC3) protein family. APOBEC3 inhibits replication of different virus families by cytosine deamination of viral DNA and a not fully characterized cytosine deamination-independent mechanism. Sheep are susceptible to small ruminant lentivirus (SRLVs) infection and contain three APOBEC3 genes encoding four proteins (A3Z1, Z2, Z3 and Z2-Z3) with yet not deeply described antiviral properties. Using sheep blood monocytes and in vitro-derived macrophages, we found that A3Z1 expression is associated with lower viral replication in this cellular type. A3Z1 transcripts may also contain spliced variants (A3Z1Tr) lacking the cytidine deaminase motif. A3Z1 exogenous expression in fully permissive fibroblast-like cells restricted SRLVs infection while A3Z1Tr allowed infection. A3Z1Tr was induced after SRLVs infection or stimulation of blood-derived macrophages with interferon gamma (IFN-γ). Interaction between truncated isoform and native A3Z1 protein was detected as well as incorporation of both proteins into virions. A3Z1 and A3Z1Tr interacted with SRLVs Vif, but this interaction was not associated with degradative properties. Similar A3Z1 truncated isoforms were also present in human and monkey cells suggesting a conserved alternative splicing regulation in primates. A3Z1-mediated retroviral restriction could be constrained by different means, including gene expression and specific alternative splicing regulation, leading to truncated protein isoforms lacking a cytidine-deaminase motif.


Assuntos
Citosina Desaminase/genética , Lentivirus/fisiologia , Replicação Viral , Processamento Alternativo/genética , Animais , Citosina Desaminase/química , Citosina Desaminase/metabolismo , Regulação da Expressão Gênica , Haplorrinos , Humanos , Interferon gama/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Motivos de Nucleotídeos/genética , Isoformas de Proteínas/genética , Ovinos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA