Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cell ; 172(5): 966-978.e12, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29474922

RESUMO

Ebola virus nucleoprotein (eNP) assembles into higher-ordered structures that form the viral nucleocapsid (NC) and serve as the scaffold for viral RNA synthesis. However, molecular insights into the NC assembly process are lacking. Using a hybrid approach, we characterized the NC-like assembly of eNP, identified novel regulatory elements, and described how these elements impact function. We generated a three-dimensional structure of the eNP NC-like assembly at 5.8 Å using electron cryo-microscopy and identified a new regulatory role for eNP helices α22-α23. Biochemical, biophysical, and mutational analyses revealed that inter-eNP contacts within α22-α23 are critical for viral NC assembly and regulate viral RNA synthesis. These observations suggest that the N terminus and α22-α23 of eNP function as context-dependent regulatory modules (CDRMs). Our current study provides a framework for a structural mechanism for NC-like assembly and a new therapeutic target.


Assuntos
Microscopia Crioeletrônica , Ebolavirus/fisiologia , Ebolavirus/ultraestrutura , Nucleocapsídeo/ultraestrutura , Nucleoproteínas/ultraestrutura , Montagem de Vírus , Modelos Biológicos , Proteínas Mutantes/química , Mutação/genética , Nucleoproteínas/química , Multimerização Proteica , Estrutura Secundária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , RNA Viral/biossíntese , RNA Viral/química , RNA Viral/metabolismo
2.
J Nat Prod ; 86(1): 182-190, 2023 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-36580354

RESUMO

Previous chemical investigation of the Irish deep-sea soft coral Duva florida led to the identification of tuaimenal A (10), a new merosesquiterpene containing a highly substituted chromene core and modest cytotoxicity against cervical cancer. Further MS/MS and NMR-guided investigation of this octocoral has resulted in the isolation and characterization of seven additional tuaimenal analogs, B-H (1-7), as well as two known A-ring aromatized steroids (8, 9), and additional tuaimenal A (10). Tuaimenals B, F, and G (1, 5, 6), bearing an oxygen at the C5 position, as well as monocyclic tuaimenal H (7), show increased cervical cancer inhibition profiles in comparison to that of 10. Tuaimenal G further displayed potent, selective cytotoxicity with an EC50 value of 0.04 µM against the C33A cell line compared to the CaSki cell line (EC50 20 µM). These data reveal the anticancer properties of tuaimenal analogs and suggest unique antiproliferation mechanisms across these secondary metabolites.


Assuntos
Antozoários , Neoplasias do Colo do Útero , Animais , Humanos , Feminino , Antozoários/química , Neoplasias do Colo do Útero/tratamento farmacológico , Espectrometria de Massas em Tandem , Florida , Linhagem Celular Tumoral
3.
J Nat Prod ; 85(5): 1315-1323, 2022 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-35549259

RESUMO

Cold water benthic environments are a prolific source of structurally diverse molecules with a range of bioactivities against human disease. Specimens of a previously chemically unexplored soft coral, Duva florida, were collected during a deep-sea cruise that sampled marine invertebrates along the Irish continental margin in 2018. Tuaimenal A (1), a cyclized merosesquiterpenoid representing a new carbon scaffold with a highly substituted chromene core, was discovered through exploration of the soft coral secondary metabolome via NMR-guided fractionation. The absolute configuration was determined through vibrational circular dichroism. Functional biochemical assays and in silico docking experiments found tuaimenal A selectively inhibits the viral main protease (3CLpro) of SARS-CoV-2.


Assuntos
Antozoários , COVID-19 , Animais , Antivirais/química , Antivirais/farmacologia , Florida , Simulação de Acoplamento Molecular , Inibidores de Proteases/farmacologia , SARS-CoV-2
4.
PLoS Pathog ; 14(1): e1006830, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29304101

RESUMO

The lentiviral protein Viral Infectivity Factor (Vif) counteracts the antiviral effects of host APOBEC3 (A3) proteins and contributes to persistent HIV infection. Vif targets A3 restriction factors for ubiquitination and proteasomal degradation by recruiting them to a multi-protein ubiquitin E3 ligase complex. Here, we describe a degradation-independent mechanism of Vif-mediated antagonism that was revealed through detailed structure-function studies of antibody antigen-binding fragments (Fabs) to the Vif complex. Two Fabs were found to inhibit Vif-mediated A3 neutralization through distinct mechanisms: shielding A3 from ubiquitin transfer and blocking Vif E3 assembly. Combined biochemical, cell biological and structural studies reveal that disruption of Vif E3 assembly inhibited A3 ubiquitination but was not sufficient to restore its packaging into viral particles and antiviral activity. These observations establish that Vif can neutralize A3 family members in a degradation-independent manner. Additionally, this work highlights the potential of Fabs as functional probes, and illuminates how Vif uses a multi-pronged approach involving both degradation dependent and independent mechanisms to suppress A3 innate immunity.


Assuntos
Antivirais/farmacologia , Citosina Desaminase/metabolismo , Fragmentos Fab das Imunoglobulinas/química , Produtos do Gene vif do Vírus da Imunodeficiência Humana/imunologia , Desaminases APOBEC , Antivirais/química , Proteínas Culina/química , Proteínas Culina/metabolismo , Citidina Desaminase , Células HEK293 , Infecções por HIV/imunologia , Infecções por HIV/terapia , Infecções por HIV/virologia , HIV-1/imunologia , HIV-1/metabolismo , Humanos , Ubiquitina/metabolismo , Ubiquitinação , Montagem de Vírus , Produtos do Gene vif do Vírus da Imunodeficiência Humana/química
5.
Biophys J ; 116(8): 1432-1445, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-30961890

RESUMO

Human immunodeficiency virus-1 viral infectivity factor (Vif) is an intrinsically disordered protein responsible for the ubiquitination of the APOBEC3 (A3) antiviral proteins. Vif folds when it binds Cullin-RING E3 ligase 5 and the transcription cofactor CBF-ß. A five-protein complex containing the substrate receptor (Vif, CBF-ß, Elongin-B, Elongin-C (VCBC)) and Cullin5 (CUL5) has a published crystal structure, but dynamics of this VCBC-CUL5 complex have not been characterized. Here, we use molecular dynamics (MD) simulations and NMR to characterize the dynamics of the VCBC complex with and without CUL5 and an A3 protein bound. Our simulations show that the VCBC complex undergoes global dynamics involving twisting and clamshell opening of the complex, whereas VCBC-CUL5 maintains a more static conformation, similar to the crystal structure. This observation from MD is supported by methyl-transverse relaxation-optimized spectroscopy NMR data, which indicates that the VCBC complex without CUL5 is dynamic on the µs-ms timescale. Our NMR data also show that the VCBC complex is more conformationally restricted when bound to the antiviral APOBEC3F (one of the A3 proteins), consistent with our MD simulations. Vif contains a flexible linker region located at the hinge of the VCBC complex, which changes conformation in conjunction with the global dynamics of the complex. Like other substrate receptors, VCBC can exist alone or in complex with CUL5 and other proteins in cells. Accordingly, the VCBC complex could be a good target for therapeutics that would inhibit full assembly of the ubiquitination complex by stabilizing an alternate VCBC conformation.


Assuntos
Proteínas Culina/química , Citidina Desaminase/química , Simulação de Dinâmica Molecular , Produtos do Gene vif do Vírus da Imunodeficiência Humana/química , Desaminases APOBEC , Subunidade beta de Fator de Ligação ao Core/química , Cristalização , Elonguina/química , Humanos , Cinética , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Relação Estrutura-Atividade , Ubiquitinação
6.
PLoS Pathog ; 9(1): e1003147, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23382680

RESUMO

Ebolaviruses, highly lethal zoonotic pathogens, possess longer genomes than most other non-segmented negative-strand RNA viruses due in part to long 5' and 3' untranslated regions (UTRs) present in the seven viral transcriptional units. To date, specific functions have not been assigned to these UTRs. With reporter assays, we demonstrated that the Zaire ebolavirus (EBOV) 5'-UTRs lack internal ribosomal entry site function. However, the 5'-UTRs do differentially regulate cap-dependent translation when placed upstream of a GFP reporter gene. Most dramatically, the 5'-UTR derived from the viral polymerase (L) mRNA strongly suppressed translation of GFP compared to a ß-actin 5'-UTR. The L 5'-UTR is one of four viral genes to possess upstream AUGs (uAUGs), and ablation of each uAUG enhanced translation of the primary ORF (pORF), most dramatically in the case of the L 5'-UTR. The L uAUG was sufficient to initiate translation, is surrounded by a "weak" Kozak sequence and suppressed pORF translation in a position-dependent manner. Under conditions where eIF2α was phosphorylated, the presence of the uORF maintained translation of the L pORF, indicating that the uORF modulates L translation in response to cellular stress. To directly address the role of the L uAUG in virus replication, a recombinant EBOV was generated in which the L uAUG was mutated to UCG. Strikingly, mutating two nucleotides outside of previously-defined protein coding and cis-acting regulatory sequences attenuated virus growth to titers 10-100-fold lower than a wild-type virus in Vero and A549 cells. The mutant virus also exhibited decreased viral RNA synthesis as early as 6 hours post-infection and enhanced sensitivity to the stress inducer thapsigargin. Cumulatively, these data identify novel mechanisms by which EBOV regulates its polymerase expression, demonstrate their relevance to virus replication and identify a potential therapeutic target.


Assuntos
RNA Polimerases Dirigidas por DNA , Ebolavirus/enzimologia , Regulação Enzimológica da Expressão Gênica , Regulação Viral da Expressão Gênica , Proteínas Virais/metabolismo , Replicação Viral/genética , Regiões 3' não Traduzidas/genética , Regiões 5' não Traduzidas/genética , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Ebolavirus/genética , Inibidores Enzimáticos/farmacologia , Humanos , Dados de Sequência Molecular , Biossíntese de Proteínas , RNA Viral/biossíntese , Tapsigargina/farmacologia , Células Vero
7.
Biochemistry ; 52(47): 8406-19, 2013 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-24067086

RESUMO

Viral protein 35 (VP35), encoded by filoviruses, is a multifunctional dsRNA binding protein that plays important roles in viral replication, innate immune evasion, and pathogenesis. The multifunctional nature of these proteins also presents opportunities to develop countermeasures that target distinct functional regions. However, functional validation and the establishment of therapeutic approaches toward such multifunctional proteins, particularly for nonenzymatic targets, are often challenging. Our previous work on filoviral VP35 proteins defined conserved basic residues located within its C-terminal dsRNA binding interferon (IFN) inhibitory domain (IID) as important for VP35 mediated IFN antagonism and viral polymerase cofactor functions. In the current study, we used a combination of structural and functional data to determine regions of Ebola virus (EBOV) VP35 (eVP35) to target for aptamer selection using SELEX. Select aptamers, representing, two distinct classes, were further characterized based on their interaction properties to eVP35 IID. These results revealed that these aptamers bind to distinct regions of eVP35 IID with high affinity (10-50 nM) and specificity. These aptamers can compete with dsRNA for binding to eVP35 and disrupt the eVP35-nucleoprotein (NP) interaction. Consistent with the ability to antagonize the eVP35-NP interaction, select aptamers can inhibit the function of the EBOV polymerase complex reconstituted by the expression of select viral proteins. Taken together, our results support the identification of two aptamers that bind filoviral VP35 proteins with high affinity and specificity and have the capacity to potentially function as filoviral VP35 protein inhibitors.


Assuntos
Antivirais/química , Aptâmeros de Nucleotídeos/química , RNA/química , Proteínas Virais Reguladoras e Acessórias/antagonistas & inibidores , Sequência de Aminoácidos , Antivirais/metabolismo , Antivirais/farmacologia , Aptâmeros de Nucleotídeos/metabolismo , Aptâmeros de Nucleotídeos/farmacologia , Ligação Competitiva , Sequência Conservada , Ebolavirus/efeitos dos fármacos , Ebolavirus/metabolismo , Doença pelo Vírus Ebola/tratamento farmacológico , Doença pelo Vírus Ebola/metabolismo , Cinética , Terapia de Alvo Molecular , Proteínas Mutantes/antagonistas & inibidores , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Conformação de Ácido Nucleico , Nucleoproteínas/metabolismo , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , RNA/metabolismo , RNA/farmacologia , RNA de Cadeia Dupla/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Técnica de Seleção de Aptâmeros , Especificidade da Espécie , Proteínas Virais Reguladoras e Acessórias/química , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/metabolismo
8.
J Immunother Cancer ; 10(7)2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35793871

RESUMO

Immunotherapies are a major breakthrough in oncology, yielding unprecedented response rates for some cancers. Especially in combination with conventional treatments or targeted agents, immunotherapeutics offer invaluable tools to improve outcomes for many patients. However, why not all patients have a favorable response remains unclear. There is an increasing appreciation of the contributions of the complex tumor microenvironment, and the tumor-immune ecosystem in particular, to treatment outcome. To date, however, there exists no immune biomarker to explain why two patients with similar clinical stage and molecular profile would have different treatment outcomes. We hypothesize that it is critical to understand both the immune and tumor states to understand how the complex system will respond to treatment. Here, we present how integrated mathematical oncology approaches can help conceptualize the effect of various immunotherapies on a patient's tumor and local immune environment, and how combinations of immunotherapy and cytotoxic therapy may be used to improve tumor response and control and limit toxicity on a per patient basis.


Assuntos
Ecossistema , Imunoterapia , Humanos , Fatores Imunológicos , Oncologia , Microambiente Tumoral
9.
STAR Protoc ; 3(1): 101030, 2022 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-34977676

RESUMO

This protocol describes the reconstitution of the filamentous Ebola virus nucleocapsid-like assembly in vitro. This is followed by solving the cryo-EM structure using helical reconstruction, and flexible fitting of the existing model into the 5.8 Å cryo-EM map. The protocol can be applied to other filamentous viral protein assemblies, particularly those with high flexibility and moderate resolution maps, which present technical challenges to model building. For complete details on the use and execution of this profile, please refer to Su et al. (2018).


Assuntos
Ebolavirus , Doença pelo Vírus Ebola , Microscopia Crioeletrônica/métodos , Ebolavirus/química , Humanos , Nucleocapsídeo/química , Montagem de Vírus
10.
J Virol ; 84(20): 10581-91, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20686031

RESUMO

The ebolavirus (EBOV) VP35 protein binds to double-stranded RNA (dsRNA), inhibits host alpha/beta interferon (IFN-α/ß) production, and is an essential component of the viral polymerase complex. Structural studies of the VP35 C-terminal IFN inhibitory domain (IID) identified specific structural features, including a central basic patch and a hydrophobic pocket, that are important for dsRNA binding and IFN inhibition. Several other conserved basic residues bordering the central basic patch and a separate cluster of basic residues, called the first basic patch, were also identified. Functional analysis of alanine substitution mutants indicates that basic residues outside the central basic patch are not required for dsRNA binding or for IFN inhibition. However, minigenome assays, which assess viral RNA polymerase complex function, identified these other basic residues to be critical for viral RNA synthesis. Of these, a subset located within the first basic patch is important for VP35-nucleoprotein (NP) interaction, as evidenced by the inability of alanine substitution mutants to coimmunoprecipitate with NP. Therefore, first basic patch residues are likely critical for replication complex formation through interactions with NP. Coimmunoprecipitation studies further demonstrate that the VP35 IID is sufficient to interact with NP and that dsRNA can modulate VP35 IID interactions with NP. Other basic residue mutations that disrupt the VP35 polymerase cofactor function do not affect interaction with NP or with the amino terminus of the viral polymerase. Collectively, these results highlight the importance of conserved basic residues from the EBOV VP35 C-terminal IID and validate the VP35 IID as a potential therapeutic target.


Assuntos
Ebolavirus/fisiologia , Proteínas Virais Reguladoras e Acessórias/química , Proteínas Virais Reguladoras e Acessórias/fisiologia , Substituição de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Ebolavirus/genética , Ebolavirus/patogenicidade , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas do Nucleocapsídeo , Nucleoproteínas/química , Nucleoproteínas/genética , Nucleoproteínas/fisiologia , Domínios e Motivos de Interação entre Proteínas , Estrutura Terciária de Proteína , RNA/genética , RNA/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Eletricidade Estática , Células Vero , Proteínas do Core Viral/química , Proteínas do Core Viral/genética , Proteínas do Core Viral/fisiologia , Proteínas Virais Reguladoras e Acessórias/genética , Virulência/genética , Virulência/fisiologia
11.
Cell Host Microbe ; 26(6): 739-747.e4, 2019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31830442

RESUMO

Primate lentiviruses encode a Vif protein that counteracts the host antiviral APOBEC3 (A3) family members. The adaptation of Vif to species-specific A3 determinants is a critical event that allowed the spillover of a lentivirus from monkey reservoirs to chimpanzees and subsequently to humans, which gave rise to HIV-1 and the acquired immune deficiency syndrome (AIDS) pandemic. How Vif-A3 protein interactions are remodeled during evolution is unclear. Here, we report a 2.94 Å crystal structure of the Vif substrate receptor complex from simian immunodeficiency virus isolated from red-capped mangabey (SIVrcm). The structure of the SIVrcm Vif complex illuminates the stage of lentiviral Vif evolution that is immediately prior to entering hominid primates. Structure-function studies reveal the adaptations that allowed SIVrcm Vif to antagonize hominid A3G. These studies show a partitioning between an evolutionarily dynamic specificity determinant and a conserved protein interacting surface on Vif that enables adaptation while maintaining protein interactions required for potent A3 antagonism.


Assuntos
Produtos do Gene vif , Vírus da Imunodeficiência Símia , Desaminase APOBEC-3G/metabolismo , Síndrome da Imunodeficiência Adquirida , Animais , Cercocebus , Cristalografia , Evolução Molecular , Produtos do Gene vif/química , Produtos do Gene vif/genética , HIV-1/genética , HIV-1/metabolismo , Hominidae , Interações Hospedeiro-Patógeno , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Doenças dos Macacos/virologia , Pan troglodytes , Primatas , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/metabolismo , Produtos do Gene vif do Vírus da Imunodeficiência Humana/química , Produtos do Gene vif do Vírus da Imunodeficiência Humana/genética , Produtos do Gene vif do Vírus da Imunodeficiência Humana/ultraestrutura
12.
Annu Rev Virol ; 4(1): 241-260, 2017 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-28961413

RESUMO

Viruses are completely dependent upon cellular machinery to support replication and have therefore developed strategies to co-opt cellular processes to optimize infection and counter host immune defenses. Many viruses, including human immunodeficiency virus type 1 (HIV-1), encode a relatively small number of genes. Viruses with limited genetic content often encode multifunctional proteins that function at multiple stages of the viral replication cycle. In this review, we discuss the functions of HIV-1 regulatory (Tat and Rev) and accessory (Vif, Vpr, Vpu, and Nef) proteins. Each of these proteins has a highly conserved primary activity; however, numerous additional activities have been attributed to these viral proteins. We explore the possibility that HIV-1 proteins leverage their multifunctional nature to alter host transcriptional networks to elicit a diverse set of cellular responses. Although these transcriptional effects appear to benefit the virus, it is not yet clear whether they are strongly selected for during viral evolution or are a ripple effect from the primary function. As our detailed knowledge of these viral proteins improves, we will undoubtedly uncover how the multifunctional nature of these HIV-1 regulatory and accessory proteins, and in particular their transcriptional functions, work to drive viral pathogenesis.


Assuntos
Genes rev , Genes tat , HIV-1/genética , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Transcrição Gênica , Proteínas Virais Reguladoras e Acessórias/metabolismo , HIV-1/química , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Proteínas do Vírus da Imunodeficiência Humana/genética , Humanos , Proteínas Virais Reguladoras e Acessórias/genética , Replicação Viral , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Produtos do Gene vif do Vírus da Imunodeficiência Humana/genética , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo
13.
Cell Rep ; 11(3): 376-89, 2015 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-25865894

RESUMO

During viral RNA synthesis, Ebola virus (EBOV) nucleoprotein (NP) alternates between an RNA-template-bound form and a template-free form to provide the viral polymerase access to the RNA template. In addition, newly synthesized NP must be prevented from indiscriminately binding to noncognate RNAs. Here, we investigate the molecular bases for these critical processes. We identify an intrinsically disordered peptide derived from EBOV VP35 (NPBP, residues 20-48) that binds NP with high affinity and specificity, inhibits NP oligomerization, and releases RNA from NP-RNA complexes in vitro. The structure of the NPBP/ΔNPNTD complex, solved to 3.7 Å resolution, reveals how NPBP peptide occludes a large surface area that is important for NP-NP and NP-RNA interactions and for viral RNA synthesis. Together, our results identify a highly conserved viral interface that is important for EBOV replication and can be targeted for therapeutic development.


Assuntos
Ebolavirus/fisiologia , Nucleoproteínas/metabolismo , RNA Viral/metabolismo , Proteínas do Core Viral/metabolismo , Calorimetria , Microscopia Crioeletrônica , Cristalografia por Raios X , Células HeLa , Doença pelo Vírus Ebola/metabolismo , Humanos , Proteínas do Nucleocapsídeo , Estrutura Quaternária de Proteína , Replicação Viral
14.
Cell Rep ; 11(8): 1236-50, 2015 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-25981045

RESUMO

HIV-1 encodes the accessory protein Vif, which hijacks a host Cullin-RING ubiquitin ligase (CRL) complex as well as the non-canonical cofactor CBFß, to antagonize APOBEC3 antiviral proteins. Non-canonical cofactor recruitment to CRL complexes by viral factors, to date, has only been attributed to HIV-1 Vif. To further study this phenomenon, we employed a comparative approach combining proteomic, biochemical, structural, and virological techniques to investigate Vif complexes across the lentivirus genus, including primate (HIV-1 and simian immunodeficiency virus macaque [SIVmac]) and non-primate (FIV, BIV, and MVV) viruses. We find that CBFß is completely dispensable for the activity of non-primate lentiviral Vif proteins. Furthermore, we find that BIV Vif requires no cofactor and that MVV Vif requires a novel cofactor, cyclophilin A (CYPA), for stable CRL complex formation and anti-APOBEC3 activity. We propose modular conservation of Vif complexes allows for potential exaptation of functions through the acquisition of non-CRL-associated host cofactors while preserving anti-APOBEC3 activity.


Assuntos
Citosina Desaminase/antagonistas & inibidores , Produtos do Gene vif/imunologia , HIV-1/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Desaminases APOBEC , Animais , Citidina Desaminase , Humanos , Ligação Proteica , Ovinos , Ubiquitina-Proteína Ligases/genética
15.
J Mol Biol ; 426(10): 2045-58, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24495995

RESUMO

The Ebola virus (EBOV) genome only encodes a single viral polypeptide with enzymatic activity, the viral large (L) RNA-dependent RNA polymerase protein. However, currently, there is limited information about the L protein, which has hampered the development of antivirals. Therefore, antifiloviral therapeutic efforts must include additional targets such as protein-protein interfaces. Viral protein 35 (VP35) is multifunctional and plays important roles in viral pathogenesis, including viral mRNA synthesis and replication of the negative-sense RNA viral genome. Previous studies revealed that mutation of key basic residues within the VP35 interferon inhibitory domain (IID) results in significant EBOV attenuation, both in vitro and in vivo. In the current study, we use an experimental pipeline that includes structure-based in silico screening and biochemical and structural characterization, along with medicinal chemistry, to identify and characterize small molecules that target a binding pocket within VP35. NMR mapping experiments and high-resolution x-ray crystal structures show that select small molecules bind to a region of VP35 IID that is important for replication complex formation through interactions with the viral nucleoprotein (NP). We also tested select compounds for their ability to inhibit VP35 IID-NP interactions in vitro as well as VP35 function in a minigenome assay and EBOV replication. These results confirm the ability of compounds identified in this study to inhibit VP35-NP interactions in vitro and to impair viral replication in cell-based assays. These studies provide an initial framework to guide development of antifiloviral compounds against filoviral VP35 proteins.


Assuntos
Antivirais/química , Coenzimas/antagonistas & inibidores , Ebolavirus/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química , Proteínas Virais Reguladoras e Acessórias/antagonistas & inibidores , Antivirais/farmacologia , Coenzimas/química , Simulação por Computador , Cristalografia por Raios X , RNA Polimerases Dirigidas por DNA/antagonistas & inibidores , RNA Polimerases Dirigidas por DNA/metabolismo , Ebolavirus/enzimologia , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas/fisiologia , Pirróis/química , Pirróis/metabolismo , Pirróis/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas Virais Reguladoras e Acessórias/química
16.
Front Microbiol ; 3: 29, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22347221

RESUMO

Aptamers generated from randomized libraries of nucleic acids have found utility in a wide variety of fields and in the clinic. Aptamers can be used to target both intracellular and extracellular components, including small molecules, proteins, cells, and viruses. With recent technological developments in stringent selection and rapid isolation strategies, it is likely that aptamers will continue to make an impact as useful tools and reagents. Although many recently developed aptamers are intended for use as therapeutic and diagnostic agents, use of aptamers for basic research, including target validation, remains an active area with high potential to impact our understanding of molecular mechanisms and for drug discovery. In this brief review, we will discuss recent aptamer discoveries, their potential role in structural virology, as well as challenges and future prospects.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa