Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 134
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Viruses ; 13(9)2021 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-34578445

RESUMO

The classical laboratory mouse strains are genetic mosaics of three Mus musculus subspecies that occupy distinct regions of Eurasia. These strains and subspecies carry infectious and endogenous mouse leukemia viruses (MLVs) that can be pathogenic and mutagenic. MLVs evolved in concert with restrictive host factors with some under positive selection, including the XPR1 receptor for xenotropic/polytropic MLVs (X/P-MLVs) and the post-entry restriction factor Fv1. Since positive selection marks host-pathogen genetic conflicts, we examined MLVs for counter-adaptations at sites that interact with XPR1, Fv1, and the CAT1 receptor for ecotropic MLVs (E-MLVs). Results describe different co-adaptive evolutionary paths within the ranges occupied by these virus-infected subspecies. The interface of CAT1, and the otherwise variable E-MLV envelopes, is highly conserved; antiviral protection is afforded by the Fv4 restriction factor. XPR1 and X/P-MLVs variants show coordinate geographic distributions, with receptor critical sites in envelope, under positive selection but with little variation in envelope and XPR1 in mice carrying P-ERVs. The major Fv1 target in the viral capsid is under positive selection, and the distribution of Fv1 alleles is subspecies-correlated. These data document adaptive, spatial and temporal, co-evolutionary trajectories at the critical interfaces of MLVs and the host factors that restrict their replication.


Assuntos
Canais de Cálcio/genética , Retrovirus Endógenos/genética , Evolução Molecular , Vírus da Leucemia Murina/genética , Proteínas/genética , Canais de Cátion TRPV/genética , Proteínas do Envelope Viral/metabolismo , Adaptação Fisiológica , Animais , Canais de Cálcio/metabolismo , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Retrovirus Endógenos/fisiologia , Interações Hospedeiro-Patógeno , Vírus da Leucemia Murina/fisiologia , Camundongos , Proteínas/metabolismo , Seleção Genética , Canais de Cátion TRPV/metabolismo , Receptor do Retrovírus Politrópico e Xenotrópico/genética , Receptor do Retrovírus Politrópico e Xenotrópico/metabolismo
2.
ACS Nano ; 15(5): 8110-8119, 2021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-33285072

RESUMO

Bioenergetic processes in nature have relied on networks of cofactors for harvesting, storing, and transforming the energy from sunlight into chemical bonds. Models mimicking the structural arrangement and functional crosstalk of the cofactor arrays are important tools to understand the basic science of natural systems and to provide guidance for non-natural functional biomaterials. Here, we report an artificial multiheme system based on a circular permutant of the tobacco mosaic virus coat protein (cpTMV). The double disk assembly of cpTMV presents a gap region sandwiched by the two C2-symmetrically related disks. Non-native bis-his coordination sites formed by the mutation of the residues in this gap region were computationally screened and experimentally tested. A cpTMV mutant Q101H was identified to create a circular assembly of 17 protein-embedded hemes. Biophysical characterization using X-ray crystallography, cyclic voltammetry, and electron paramagnetic resonance (EPR) suggested both structural and functional similarity to natural multiheme cytochrome c proteins. This protein framework offers many further engineering opportunities for tuning the redox properties of the cofactors and incorporating non-native components bearing varied porphyrin structures and metal centers. Emulating the electron transfer pathways in nature using a tunable artificial system can contribute to the development of photocatalytic materials and bioelectronics.


Assuntos
Metaloporfirinas , Vírus do Mosaico do Tabaco , Proteínas do Capsídeo/genética , Compostos Orgânicos , Análise Serial de Proteínas , Proteínas , Vírus do Mosaico do Tabaco/genética
3.
Proc Natl Acad Sci U S A ; 115(45): 11519-11524, 2018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30333189

RESUMO

The host factor protein TRIM5α plays an important role in restricting the host range of HIV-1, interfering with the integrity of the HIV-1 capsid. TRIM5 triggers an antiviral innate immune response by functioning as a capsid pattern recognition receptor, although the precise mechanism by which the restriction is imposed is not completely understood. Here we used an integrated magic-angle spinning nuclear magnetic resonance and molecular dynamics simulations approach to characterize, at atomic resolution, the dynamics of the capsid's hexameric and pentameric building blocks, and the interactions with TRIM5α in the assembled capsid. Our data indicate that assemblies in the presence of the pentameric subunits are more rigid on the microsecond to millisecond timescales than tubes containing only hexamers. This feature may be of key importance for controlling the capsid's morphology and stability. In addition, we found that TRIM5α binding to capsid induces global rigidification and perturbs key intermolecular interfaces essential for higher-order capsid assembly, with structural and dynamic changes occurring throughout the entire CA polypeptide chain in the assembly, rather than being limited to a specific protein-protein interface. Taken together, our results suggest that TRIM5α uses several mechanisms to destabilize the capsid lattice, ultimately inducing its disassembly. Our findings add to a growing body of work indicating that dynamic allostery plays a pivotal role in capsid assembly and HIV-1 infectivity.


Assuntos
Proteínas do Capsídeo/metabolismo , Capsídeo/metabolismo , HIV-1/metabolismo , Proteínas/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Capsídeo/química , Capsídeo/ultraestrutura , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , HIV-1/genética , HIV-1/ultraestrutura , Humanos , Macaca mulatta , Espectroscopia de Ressonância Magnética/métodos , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas/química , Proteínas/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Ubiquitina-Proteína Ligases
4.
Methods Mol Biol ; 1798: 195-201, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29868961

RESUMO

The use of antibody conjugates for biomedical applications has garnered increased attention due to the ability of antibodies to specifically engage targets of interest. Despite these appealing qualities, the preparation of antibody-protein conjugates remains challenging. Here we detail an approach to attaching targeting antibodies to proteins of interest that combines advances in genetic code expansion and an efficient bioconjugation strategy. As an example, we prepare bacteriophage MS2 viral capsids bearing antibodies on their surfaces for applications in molecular targeting. This technique provides a modular framework to easily prepare antibody-MS2 conjugates in an efficient manner, even at low concentrations of the reacting biomolecules.


Assuntos
Anticorpos/química , Fenilalanina/análogos & derivados , Proteínas/química , Anticorpos/genética , Capsídeo/química , Capsídeo/metabolismo , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Códon , Levivirus , Fenilalanina/química , Proteínas/genética
5.
J Virol ; 92(10)2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29491156

RESUMO

Cytidine deaminases inhibit replication of a broad range of DNA viruses by deaminating cytidines on single-stranded DNA (ssDNA) to generate uracil. While several lines of evidence have revealed hepatitis B virus (HBV) genome editing by deamination, it is still unclear which nucleic acid intermediate of HBV is modified. Hepatitis B virus has a relaxed circular double-stranded DNA (rcDNA) genome that is reverse transcribed within virus cores from a RNA template. The HBV genome also persists as covalently closed circular DNA (cccDNA) in the nucleus of an infected cell. In the present study, we found that in HBV-producing HepAD38 and HepG2.2.15 cell lines, endogenous cytidine deaminases edited 10 to 25% of HBV rcDNA genomes, asymmetrically with almost all mutations on the 5' half of the minus strand. This region corresponds to the last half of the minus strand to be protected by plus-strand synthesis. Within this half of the genome, the number of mutations peaks in the middle. Overexpressed APOBEC3A and APOBEC3G could be packaged in HBV capsids but did not change the amount or distribution of mutations. We found no deamination on pregenomic RNA (pgRNA), indicating that an intact genome is encapsidated and deaminated during or after reverse transcription. The deamination pattern suggests a model of rcDNA synthesis in which pgRNA and then newly synthesized minus-sense single-stranded DNA are protected from deaminase by interaction with the virus capsid; during plus-strand synthesis, when enough dsDNA has been synthesized to displace the remaining minus strand from the capsid surface, the single-stranded DNA becomes deaminase sensitive.IMPORTANCE Host-induced mutation of the HBV genome by APOBEC proteins may be a path to clearing the virus. We examined cytidine-to-thymidine mutations in the genomes of HBV particles grown in the presence or absence of overexpressed APOBEC proteins. We found that genomes were subjected to deamination activity during reverse transcription, which takes place within the virus capsid. These observations provide a direct insight into the mechanics of reverse transcription, suggesting that newly synthesized dsDNA displaces ssDNA from the capsid walls, making the ssDNA accessible to deaminase activity.


Assuntos
Desaminase APOBEC-3G/metabolismo , Citidina Desaminase/metabolismo , DNA Viral/metabolismo , Vírus da Hepatite B/genética , Proteínas/metabolismo , RNA Viral/metabolismo , Transcrição Reversa/genética , Capsídeo/metabolismo , Linhagem Celular Tumoral , Empacotamento do DNA/genética , DNA Circular/metabolismo , DNA Viral/genética , Desaminação/genética , Genoma Viral/genética , Células Hep G2 , Vírus da Hepatite B/enzimologia , Humanos , Mutação/genética , RNA Viral/genética
6.
J Virol ; 92(5)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29237846

RESUMO

Rhesus TRIM5α (rhTRIM5α) potently restricts replication of human immunodeficiency virus type 1 (HIV-1). Restriction is mediated through direct binding of the C-terminal B30.2 domain of TRIM5α to the assembled HIV-1 capsid core. This host-pathogen interaction involves multiple capsid molecules within the hexagonal HIV-1 capsid lattice. However, the molecular details of this interaction and the precise site at which the B30.2 domain binds remain largely unknown. The human orthologue of TRIM5α (hsTRIM5α) fails to block infection by HIV-1 both in vivo and in vitro This is thought to be due to differences in binding to the capsid lattice. To map the species-specific binding surface on the HIV-1 capsid lattice, we used microscale thermophoresis and dual-focus fluorescence correlation spectroscopy to measure binding affinity of rhesus and human TRIM5α B30.2 domains to a series of HIV-1 capsid variants that mimic distinct capsid arrangements at each of the symmetry axes of the HIV-1 capsid lattice. These surrogates include previously characterized capsid oligomers, as well as a novel chemically cross-linked capsid trimer that contains cysteine substitutions near the 3-fold axis of symmetry. The results demonstrate that TRIM5α binding involves multiple capsid molecules along the 2-fold and 3-fold interfaces between hexamers and indicate that the binding interface at the 3-fold axis contributes to the well-established differences in restriction potency between TRIM5α orthologues.IMPORTANCE TRIM5α is a cellular protein that fends off infection by retroviruses through binding to the viruses' protein shell surrounding its genetic material. This shell is composed of several hundred capsid proteins arranged in a honeycomb-like hexagonal pattern that is conserved across retroviruses. By binding to the complex lattice formed by multiple capsid proteins, rather than to a single capsid monomer, TRIM5α restriction activity persists despite the high mutation rate in retroviruses such as HIV-1. In rhesus monkeys, but not in humans, TRIM5α confers resistance to HIV-1. By measuring the binding of human and rhesus TRIM5α to a series of engineered HIV-1 capsid mimics of distinct capsid lattice interfaces, we reveal the HIV-1 capsid surface critical for species-specific binding by TRIM5α.


Assuntos
Proteínas do Capsídeo/química , Proteínas de Transporte/química , HIV-1/química , Proteínas/química , Animais , Fatores de Restrição Antivirais , Proteínas do Capsídeo/genética , Cristalografia por Raios X , Ciclofilina A/química , Ciclofilina A/genética , HIV-1/genética , HIV-1/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Macaca mulatta , Modelos Moleculares , Mutação , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes , Células Sf9 , Especificidade da Espécie , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases
7.
J Virol ; 92(4)2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29187540

RESUMO

Restriction factors are intrinsic cellular defense proteins that have evolved to block microbial infections. Retroviruses such as HIV-1 are restricted by TRIM5 proteins, which recognize the viral capsid shell that surrounds, organizes, and protects the viral genome. TRIM5α uses a SPRY domain to bind capsids with low intrinsic affinity (KD of >1 mM) and therefore requires higher-order assembly into a hexagonal lattice to generate sufficient avidity for productive capsid recognition. TRIMCyp, on the other hand, binds HIV-1 capsids through a cyclophilin A domain, which has a well-defined binding site and higher affinity (KD of ∼10 µM) for isolated capsid subunits. Therefore, it has been argued that TRIMCyp proteins have dispensed with the need for higher-order assembly to function as antiviral factors. Here, we show that, consistent with its high degree of sequence similarity with TRIM5α, the TRIMCyp B-box 2 domain shares the same ability to self-associate and facilitate assembly of a TRIMCyp hexagonal lattice that can wrap about the HIV-1 capsid. We also show that under stringent experimental conditions, TRIMCyp-mediated restriction of HIV-1 is indeed dependent on higher-order assembly. Both forms of TRIM5 therefore use the same mechanism of avidity-driven capsid pattern recognition.IMPORTANCE Rhesus macaques and owl monkeys are highly resistant to HIV-1 infection due to the activity of TRIM5 restriction factors. The rhesus macaque TRIM5α protein blocks HIV-1 through a mechanism that requires self-assembly of a hexagonal TRIM5α lattice around the invading viral core. Lattice assembly amplifies very weak interactions between the TRIM5α SPRY domain and the HIV-1 capsid. Assembly also promotes dimerization of the TRIM5α RING E3 ligase domain, resulting in synthesis of polyubiquitin chains that mediate downstream steps of restriction. In contrast to rhesus TRIM5α, the owl monkey TRIM5 homolog, TRIMCyp, binds isolated HIV-1 CA subunits much more tightly through its cyclophilin A domain and therefore was thought to act independently of higher-order assembly. Here, we show that TRIMCyp shares the assembly properties of TRIM5α and that both forms of TRIM5 use the same mechanism of hexagonal lattice formation to promote viral recognition and restriction.


Assuntos
Proteínas do Capsídeo/metabolismo , Capsídeo/metabolismo , Proteínas de Transporte/metabolismo , HIV-1/genética , Proteínas/metabolismo , Sequência de Aminoácidos , Animais , Fatores de Restrição Antivirais , Aotidae , Proteínas do Capsídeo/ultraestrutura , Proteínas de Transporte/genética , HIV-1/metabolismo , Células HeLa , Humanos , Macaca mulatta , Domínios Proteicos , Multimerização Proteica , Proteínas/genética , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
8.
J Virol ; 92(1)2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29046456

RESUMO

Efficient antiviral immunity requires interference with virus replication at multiple layers targeting diverse steps in the viral life cycle. We describe here a novel flavivirus inhibition mechanism that results in interferon-mediated obstruction of tick-borne encephalitis virus particle assembly and involves release of malfunctioning membrane-associated capsid (C) particles. This mechanism is controlled by the activity of the interferon-induced protein viperin, a broad-spectrum antiviral interferon-stimulated gene. Through analysis of the viperin-interactome, we identified the Golgi brefeldin A-resistant guanine nucleotide exchange factor 1 (GBF1) as the cellular protein targeted by viperin. Viperin-induced antiviral activity, as well as C-particle release, was stimulated by GBF1 inhibition and knockdown and reduced by elevated levels of GBF1. Our results suggest that viperin targets flavivirus virulence by inducing the secretion of unproductive noninfectious virus particles via a GBF1-dependent mechanism. This as-yet-undescribed antiviral mechanism allows potential therapeutic intervention.IMPORTANCE The interferon response can target viral infection on almost every level; however, very little is known about the interference of flavivirus assembly. We show here that interferon, through the action of viperin, can disturb the assembly of tick-borne encephalitis virus. The viperin protein is highly induced after viral infection and exhibit broad-spectrum antiviral activity. However, the mechanism of action is still elusive and appears to vary between the different viruses, indicating that cellular targets utilized by several viruses might be involved. In this study, we show that viperin induces capsid particle release by interacting and inhibiting the function of the cellular protein Golgi brefeldin A-resistant guanine nucleotide exchange factor 1 (GBF1). GBF1 is a key protein in the cellular secretory pathway and is essential in the life cycle of many viruses, also targeted by viperin, implicating GBF1 as a novel putative drug target.


Assuntos
Infecções por Flavivirus/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Interferon Tipo I/farmacologia , Proteínas/metabolismo , Células A549 , Animais , Proteínas do Capsídeo/metabolismo , Chlorocebus aethiops , Flavivirus/efeitos dos fármacos , Flavivirus/patogenicidade , Infecções por Flavivirus/tratamento farmacológico , Infecções por Flavivirus/virologia , Regulação da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/genética , Células HEK293 , Células HeLa , Humanos , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Células Vero , Virulência , Montagem de Vírus/efeitos dos fármacos
9.
J Virol ; 91(10)2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28275184

RESUMO

Human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) replication in human cells is restricted at early postentry steps by host inhibitory factors. We previously described and characterized an early-phase restriction of HIV-1 and -2 replication in human cell lines, primary macrophages, and peripheral blood mononuclear cells. The restriction was termed lentiviral restriction 2 (Lv2). The viral determinants of Lv2 susceptibility mapped to the HIV-2 envelope (Env) and capsid (CA). We subsequently reported a whole-genome small interfering RNA screening for factors involved in HIV that identified RNA-associated early-stage antiviral factor (REAF). Using HIV-2 chimeras of susceptible and nonsusceptible viruses, we show here that REAF is a major component of the previously described Lv2 restriction. Further studies of the viral CA demonstrate that the CA mutation I73V (previously called I207V), a potent determinant for HIV-2, is a weak determinant of susceptibility for HIV-1. More potent CA determinants for HIV-1 REAF restriction were identified at P38A, N74D, G89V, and G94D. These results firmly establish that in HIV-1, CA is a strong determinant of susceptibility to Lv2/REAF. Similar to HIV-2, HIV-1 Env can rescue sensitive CAs from restriction. We conclude that REAF is a major component of the previously described Lv2 restriction.IMPORTANCE Measures taken by the host cell to combat infection drive the evolution of pathogens to counteract or sidestep them. The study of such virus-host conflicts can point to possible weaknesses in the arsenal of viruses and may lead to the rational design of antiviral agents. Here we describe our discovery that the host restriction factor REAF fulfills the same criteria previously used to describe lentiviral restriction (Lv2). We show that, like the HIV-2 CA, the CA of HIV-1 is a strong determinant of Lv2/REAF susceptibility. We illustrate how HIV counteracts Lv2/REAF by using an envelope with alternative routes of entry into cells.


Assuntos
Imunidade Inata , Proteínas/metabolismo , Replicação Viral , Animais , Proteínas do Capsídeo , Linhagem Celular , Produtos do Gene env/genética , Genoma Viral , HIV-1/genética , HIV-2/genética , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Leucócitos Mononucleares/virologia , RNA Interferente Pequeno
10.
Chem Asian J ; 11(20): 2814-2828, 2016 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-27432619

RESUMO

Materials science is beginning to focus on biotemplation, and in support of that trend, it is realized that protein cages-proteins that assemble from multiple monomers into architectures with hollow interiors-can instill a number of unique advantages to nanomaterials. In addition, the structural and functional plasticity of many protein-cage systems permits their engineering for specific applications. In this review, the most commonly used viral and non-viral protein cages, which exhibit a wide diversity of size, functionality, and chemical and thermal stabilities, are described. Moreover, how they have been exploited for nanomaterial and nanotechnology applications is summarized.


Assuntos
Nanoestruturas/química , Engenharia de Proteínas , Proteínas/química , Bacteriófagos/metabolismo , Bromovirus/metabolismo , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Comovirus/metabolismo , Ferritinas/química , Ferritinas/metabolismo , Proteínas de Choque Térmico Pequenas/química , Proteínas de Choque Térmico Pequenas/metabolismo , Proteínas/metabolismo , Propriedades de Superfície , Vírus do Mosaico do Tabaco/metabolismo
11.
Retrovirology ; 13(1): 42, 2016 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-27342974

RESUMO

BACKGROUND: The mouse protein Fv1 is a factor that can confer resistance to retroviral infection. The two major Fv1 alleles from laboratory mice, Fv1 (n) and Fv1 (b) , restrict infection by different murine leukaemia viruses (MLVs). Fv1(n) restricts B-tropic MLV, but not N-tropic MLV or NB-tropic MLV. In cells expressing Fv1(b) at natural levels, only N-MLV is restricted, however restriction of NB-MLV and partial restriction of B-MLV were observed when recombinant Fv1(b) was expressed from an MLV promoter in Fv1 null Mus dunni tail fibroblast cells. To investigate the relationship between expression level and restriction specificity we have developed new retroviral delivery vectors which allow inducible expression of Fv1, and yet allow sufficient production of fluorescent reporter proteins for analysis in our FACS-based restriction assay. RESULTS: We demonstrated that at concentrations close to the endogenous expression level, Fv1(b) specifically restricts only N-MLV, but restriction of NB-MLV, and to a lesser extent B-MLV, could be gained by increasing the protein level of Fv1(b). By contrast, we found that even when Fv1(n) is expressed at very high levels, no significant inhibition of N-MLV or NB-MLV could be observed. Study of Fv1 mutants using this assay led to the identification of determinants for N/B tropism at an expression level close to that of endogenous Fv1(n) and Fv1(b). We also compared the recently described restriction activities of wild mice Fv1 proteins directed against non-MLV retroviruses when expressed at different levels. Fv1 from M. spretus restricted N-MLV, B-MLV and equine infectious anaemia virus equally even at low concentrations, while Fv1 from M. macedonicus showed even stronger restriction against equine infectious anaemia virus than to N-MLV. Restriction of feline foamy virus by Fv1 of M. caroli occurred at levels equivalent to MLV restriction. CONCLUSIONS: Our data indicate that for some but not all Fv1 proteins, gain of restriction activities could be achieved by increasing the expression level of Fv1. However such a concentration dependent effect is not seen with most Fv1s and cannot explain the recently reported activities against non-MLVs. It will be interesting to examine whether overexpression of other capsid binding restriction factors such as TRIM5α or Mx2 result in novel restriction specificities.


Assuntos
Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/fisiologia , Proteínas/genética , Proteínas/metabolismo , Replicação Viral , Animais , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Vetores Genéticos , Humanos , Camundongos , Mutação
12.
Biochem Biophys Res Commun ; 457(3): 295-9, 2015 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-25576866

RESUMO

Apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like (APOBEC) proteins are cellular DNA/RNA-editing enzymes that play pivotal roles in the innate immune response to viral infection. APOBEC3 (A3) proteins were reported to hypermutate the genome of human papillomavirus 16 (HPV16), the causative agent of cervical cancer. However, hypermutation did not affect viral DNA maintenance, leaving the exact role of A3 against HPV infection elusive. Here we examine whether A3 proteins affect the virion assembly using an HPV16 pseudovirion (PsV) production system, in which PsVs are assembled from its capsid proteins L1/L2 encapsidating a reporter plasmid in 293FT cells. We found that co-expression of A3A or A3C in 293FT cells greatly reduced the infectivity of PsV. The reduced infectivity of PsV assembled in the presence of A3A, but not A3C, was attributed to the decreased copy number of the encapsidated reporter plasmid. On the other hand, A3C, but not A3A, efficiently bound to L1 in co-immunoprecipitation assays, which suggests that this physical interaction may lead to reduced infectivity of PsV assembled in the presence of A3C. These results provide mechanistic insights into A3s' inhibitory effects on the assembly phase of the HPV16 virion.


Assuntos
Citidina Desaminase/fisiologia , Papillomavirus Humano 16/patogenicidade , Proteínas/fisiologia , Proteínas do Capsídeo/fisiologia , Citidina Desaminase/genética , Feminino , Genoma Viral , Células HEK293 , Interações Hospedeiro-Patógeno , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/fisiologia , Humanos , Proteínas Oncogênicas Virais/fisiologia , Ligação Proteica , Proteínas/genética , Vírion/genética , Vírion/patogenicidade , Vírion/fisiologia , Virulência , Montagem de Vírus
13.
Microbes Infect ; 16(11): 936-44, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25195168

RESUMO

TRIM5α is a potent anti-retroviral factor that interacts with viral capsid (CA) in a species-specific manner. Recently, we and others reported generation of two distinct HIV-1 CAs that effectively overcome rhesus TRIM5α-imposed species barrier. In this study, to directly compare the effect of different mutations in the two HIV-1 CAs on evasion from macaque TRIM5-restriction, we newly generated macaque-tropic HIV-1 (HIV-1mt) proviral clones carrying the distinct CAs in the same genomic backbone, and examined their replication abilities in macaque TRIM5-overexpressing human cells and in rhesus cells. Comparative analysis of amino acid sequences and homology modeling-based structures revealed that, while both CAs gained some mutated amino acids with similar physicochemical properties, their overall appearances of N-terminal domains were different. Experimentally, the two CAs exhibited incomplete TRIM5α-resistance relative to SIVmac239 CA and different degrees of susceptibility to various TRIM5 proteins. Finally, two HIV-1mt clones carrying a different combination of the CA mutations were found to grow to a comparable extent in established and primary rhesus cells. Our data show that there could be some distinct CA patterns to confer significant TRIM5-resistance on HIV-1.


Assuntos
Substituição de Aminoácidos , Infecções por HIV/imunologia , HIV-1/fisiologia , Proteínas/metabolismo , Tropismo Viral , Replicação Viral , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética , Animais , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Humanos , Macaca mulatta , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Ubiquitina-Proteína Ligases , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo
14.
Bioconjug Chem ; 25(9): 1620-5, 2014 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-25134017

RESUMO

The size, well-defined structure, and relatively high folding energies of most proteins allow them to recognize disease-relevant receptors that present a challenge to small molecule reagents. While multiple challenges must be overcome in order to fully exploit the use of protein reagents in basic research and medicine, perhaps the greatest challenge is their intracellular delivery to a particular diseased cell. Here, we describe the genetic and enzymatic manipulation of prostate cancer cell-penetrating M13 bacteriophage to generate nanocarriers for the intracellular delivery of functional exogenous proteins to a human prostate cancer cell line.


Assuntos
Bacteriófago M13/genética , Bacteriófago M13/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Engenharia Genética , Espaço Intracelular/metabolismo , Nanoestruturas , Neoplasias da Próstata/patologia , Proteínas/metabolismo , Sequência de Aminoácidos , Bacteriófago M13/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular Tumoral , Humanos , Masculino , Dados de Sequência Molecular , Neoplasias da Próstata/metabolismo , Proteínas/genética
15.
Proc Natl Acad Sci U S A ; 111(26): 9609-14, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24979782

RESUMO

Restriction factors (RFs) form important components of host defenses to retroviral infection. The Fv1, Trim5α, and TrimCyp RFs contain N-terminal dimerization and C-terminal specificity domains that target assembled retroviral capsid (CA) proteins enclosing the viral core. However, the molecular detail of the interaction between RFs and their CA targets is unknown. Therefore, we have determined the crystal structure of the B-box and coiled-coil (BCC) region from Trim5α and used small-angle X-ray scattering to examine the solution structure of Trim5α BCC, the dimerization domain of Fv1 (Fv1Ntd), and the hybrid restriction factor Fv1Cyp comprising Fv1NtD fused to the HIV-1 binding protein Cyclophilin A (CypA). These data reveal that coiled-coil regions of Fv1 and Trim5α form extended antiparallel dimers. In Fv1Cyp, two CypA moieties are located at opposing ends, creating a molecule with a dumbbell appearance. In Trim5α, the B-boxes are located at either end of the coiled-coil, held in place by interactions with a helical motif from the L2 region of the opposing monomer. A comparative analysis of Fv1Cyp and CypA binding to a preformed HIV-1 CA lattice reveals how RF dimerization enhances the affinity of interaction through avidity effects. We conclude that the antiparallel organization of the NtD regions of Fv1 and Trim5α dimers correctly positions C-terminal specificity and N-terminal effector domains and facilitates stable binding to adjacent CA hexamers in viral cores.


Assuntos
Capsídeo/metabolismo , HIV-1/metabolismo , Modelos Moleculares , Muramidase/química , Proteínas/química , Internalização do Vírus , Sequência de Aminoácidos , Animais , Bacteriófago T4/enzimologia , Sequência de Bases , Cromatografia em Gel , Cristalização , Dimerização , Escherichia coli , Modelos Lineares , Macaca mulatta , Microscopia Eletrônica , Dados de Sequência Molecular , Conformação Proteica , Proteínas/genética , Proteínas/metabolismo , Proteínas Recombinantes de Fusão/genética , Espalhamento a Baixo Ângulo , Análise de Sequência de DNA , Ressonância de Plasmônio de Superfície , Ubiquitina-Proteína Ligases , Difração de Raios X
16.
ACS Nano ; 8(5): 4166-76, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24708510

RESUMO

The fully de novo design of protein building blocks for self-assembling as functional nanoparticles is a challenging task in emerging nanomedicines, which urgently demand novel, versatile, and biologically safe vehicles for imaging, drug delivery, and gene therapy. While the use of viruses and virus-like particles is limited by severe constraints, the generation of protein-only nanocarriers is progressively reachable by the engineering of protein-protein interactions, resulting in self-assembling functional building blocks. In particular, end-terminal cationic peptides drive the organization of structurally diverse protein species as regular nanosized oligomers, offering promise in the rational engineering of protein self-assembling. However, the in vivo stability of these constructs, being a critical issue for their medical applicability, needs to be assessed. We have explored here if the cross-molecular contacts between protein monomers, generated by end-terminal cationic peptides and oligohistidine tags, are stable enough for the resulting nanoparticles to overcome biological barriers in assembled form. The analyses of renal clearance and biodistribution of several tagged modular proteins reveal long-term architectonic stability, allowing systemic circulation and tissue targeting in form of nanoparticulate material. This observation fully supports the value of the engineered of protein building blocks addressed to the biofabrication of smart, robust, and multifunctional nanoparticles with medical applicability that mimic structure and functional capabilities of viral capsids.


Assuntos
Nanomedicina/métodos , Nanopartículas/química , Proteínas/química , Animais , Capsídeo/química , Cátions , Sistemas de Liberação de Medicamentos , Feminino , Engenharia Genética , Proteínas de Fluorescência Verde/química , Histidina/química , Humanos , Ligação de Hidrogênio , Rim/metabolismo , Ligantes , Camundongos , Camundongos Nus , Microscopia de Força Atômica , Microscopia Eletrônica de Transmissão , Peptídeos/química , Ligação Proteica , Mapeamento de Interação de Proteínas , Eletricidade Estática
17.
Mol Biosyst ; 10(4): 724-7, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24469550

RESUMO

The mono-site mutations of the absolutely conserved residues, (464)LGR(466), in the α-helix 5 (h5) of HPV16 L1 completely disrupted the pentamer formation. The implication of this finding is the potential usage of a h5-like peptide as the reagent to interfere with the pentamer formation and stability as an anti-HPV reagent.


Assuntos
Proteínas do Capsídeo/genética , Proteínas do Capsídeo/ultraestrutura , Papillomavirus Humano 16/genética , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/ultraestrutura , Proteínas/metabolismo , Substituição de Aminoácidos , Dicroísmo Circular , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação , Vacinas contra Papillomavirus , Peptídeos , Estrutura Secundária de Proteína/genética , Montagem de Vírus/genética
18.
Proc Natl Acad Sci U S A ; 110(52): 21189-94, 2013 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-24324167

RESUMO

Genes encoding plant nucleotide-binding leucine-rich repeat (NB-LRR) proteins confer dominant resistance to diverse pathogens. The wild-type potato NB-LRR protein Rx confers resistance against a single strain of potato virus X (PVX), whereas LRR mutants protect against both a second PVX strain and the distantly related poplar mosaic virus (PopMV). In one of the Rx mutants there was a cost to the broad-spectrum resistance because the response to PopMV was transformed from a mild disease on plants carrying wild-type Rx to a trailing necrosis that killed the plant. To explore the use of secondary mutagenesis to eliminate this cost of broad-spectrum resistance, we performed random mutagenesis of the N-terminal domains of this broad-recognition version of Rx and isolated four mutants with a stronger response against the PopMV coat protein due to enhanced activation sensitivity. These mutations are located close to the nucleotide-binding pocket, a highly conserved structure that likely controls the "switch" between active and inactive NB-LRR conformations. Stable transgenic plants expressing one of these versions of Rx are resistant to the strains of PVX and the PopMV that previously caused trailing necrosis. We conclude from this work that artificial evolution of NB-LRR disease resistance genes in crops can be enhanced by modification of both activation and recognition phases, to both accentuate the positive and eliminate the negative aspects of disease resistance.


Assuntos
Engenharia Genética/métodos , Imunidade Inata/genética , Proteínas de Plantas/genética , Proteínas/genética , Agricultura/métodos , Agrobacterium tumefaciens , Substituição de Aminoácidos/genética , Western Blotting , Proteínas do Capsídeo/genética , Carlavirus/genética , Genes vpr/genética , Proteínas de Repetições Ricas em Leucina , Proteínas de Plantas/imunologia , Plantas Geneticamente Modificadas , Proteínas/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , /virologia
19.
BMC Biotechnol ; 13: 79, 2013 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-24073829

RESUMO

BACKGROUND: Consistent progress in the development of bacteriophage lambda display platform as an alternative to filamentous phage display system was achieved in the recent years. The lambda phage has been engineered to display efficiently multiple copies of peptides or even large protein domains providing a powerful tool for screening libraries of peptides, proteins and cDNA. RESULTS: In the present work we describe an original method for dual display of large proteins on the surface of lambda particles. An anti-CEA single-chain antibody fragment and green fluorescent protein or alkaline phosphatase were simultaneously displayed by engineering both gpD and gpV lambda proteins. CONCLUSIONS: Here we show that such modified phage particles can be used for the detection of target molecules in vitro and in vivo. Dual expression of functional moieties on the surface of the lambda phage might open the way to generation of a new class of diagnostic and therapeutic targeted nanoparticles.


Assuntos
Bacteriófago lambda/genética , Proteínas/química , Proteínas/genética , Fosfatase Alcalina/metabolismo , Animais , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Linhagem Celular , Clonagem Molecular , DNA Complementar/química , DNA Complementar/genética , Escherichia coli/genética , Vetores Genéticos , Glicoproteínas/química , Glicoproteínas/genética , Proteínas de Fluorescência Verde/genética , Células HT29 , Humanos , Camundongos , Camundongos Nus , Microscopia Confocal , Nanopartículas/química , Biblioteca de Peptídeos , Plasmídeos/genética , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
20.
J Mol Biol ; 425(24): 5032-44, 2013 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-23886867

RESUMO

The restriction factor TRIM5α binds to the capsid protein of the retroviral core and blocks retroviral replication. The affinity of TRIM5α for the capsid is a major host tropism determinant of HIV and other primate immunodeficiency viruses, but the molecular interface involved in this host-pathogen interaction remains poorly characterized. Here we use NMR spectroscopy to investigate binding of the rhesus TRIM5α SPRY domain to a selection of HIV capsid constructs. The data are consistent with a model in which one SPRY domain interacts with more than one capsid monomer within the assembled retroviral core. The highly mobile SPRY v1 loop appears to span the gap between neighboring capsid hexamers making interhexamer contacts critical for restriction. The interaction interface is extensive, involves mobile loops and multiple epitopes, and lacks interaction hot spots. These properties, which may enhance resistance of TRIM5α to capsid mutations, result in relatively low affinity of the individual SPRY domains for the capsid, and the TRIM5α-mediated restriction depends on the avidity effect arising from the oligomerization of TRIM5α.


Assuntos
Proteínas do Capsídeo/imunologia , Capsídeo/imunologia , Epitopos/imunologia , HIV-1/imunologia , Proteínas/metabolismo , Sequência de Aminoácidos , Animais , Capsídeo/química , Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Epitopos/análise , Epitopos/metabolismo , HIV-1/genética , HIV-1/metabolismo , Interações Hospedeiro-Patógeno , Macaca mulatta , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Ligação Proteica , Conformação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Proteínas/genética , Proteínas/imunologia , Alinhamento de Sequência , Ubiquitina-Proteína Ligases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA