Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 15(2)2023 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-36851772

RESUMO

The SARS-CoV-2 pandemic has again shown that structural biology plays an important role in understanding biological mechanisms and exploiting structural data for therapeutic interventions. Notably, previous work on SARS-related glycoproteins has paved the way for the rapid structural determination of the SARS-CoV-2 S glycoprotein, which is the main target for neutralizing antibodies. Therefore, all vaccine approaches aimed to employ S as an immunogen to induce neutralizing antibodies. Like all enveloped virus glycoproteins, SARS-CoV-2 S native prefusion trimers are in a metastable conformation, which primes the glycoprotein for the entry process via membrane fusion. S-mediated entry is associated with major conformational changes in S, which can expose many off-target epitopes that deviate vaccination approaches from the major aim of inducing neutralizing antibodies, which mainly target the native prefusion trimer conformation. Here, we review the viral glycoprotein stabilization methods developed prior to SARS-CoV-2, and applied to SARS-CoV-2 S, in order to stabilize S in the prefusion conformation. The importance of structure-based approaches is highlighted by the benefits of employing stabilized S trimers versus non-stabilized S in vaccines with respect to their protective efficacy.


Assuntos
COVID-19 , Humanos , COVID-19/prevenção & controle , SARS-CoV-2 , Anticorpos Neutralizantes , Epitopos , Glicoproteínas
2.
Nat Struct Mol Biol ; 30(1): 81-90, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36604498

RESUMO

The endosomal sorting complex required for transport (ESCRT) is a highly conserved protein machinery that drives a divers set of physiological and pathological membrane remodeling processes. However, the structural basis of ESCRT-III polymers stabilizing, constricting and cleaving negatively curved membranes is yet unknown. Here we present cryo-EM structures of membrane-coated CHMP2A-CHMP3 filaments from Homo sapiens of two different diameters at 3.3 and 3.6 Å resolution. The structures reveal helical filaments assembled by CHMP2A-CHMP3 heterodimers in the open ESCRT-III conformation, which generates a partially positive charged membrane interaction surface, positions short N-terminal motifs for membrane interaction and the C-terminal VPS4 target sequence toward the tube interior. Inter-filament interactions are electrostatic, which may facilitate filament sliding upon VPS4-mediated polymer remodeling. Fluorescence microscopy as well as high-speed atomic force microscopy imaging corroborate that VPS4 can constrict and cleave CHMP2A-CHMP3 membrane tubes. We therefore conclude that CHMP2A-CHMP3-VPS4 act as a minimal membrane fission machinery.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte , Polímeros , Humanos , Complexos Endossomais de Distribuição Requeridos para Transporte/química , Polímeros/metabolismo , Proteínas de Transporte/metabolismo , Transporte Proteico
3.
Cell Rep Med ; 3(2): 100528, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35233549

RESUMO

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has caused an ongoing global health crisis. Here, we present as a vaccine candidate synthetic SARS-CoV-2 spike (S) glycoprotein-coated lipid vesicles that resemble virus-like particles. Soluble S glycoprotein trimer stabilization by formaldehyde cross-linking introduces two major inter-protomer cross-links that keep all receptor-binding domains in the "down" conformation. Immunization of cynomolgus macaques with S coated onto lipid vesicles (S-LVs) induces high antibody titers with potent neutralizing activity against the vaccine strain, Alpha, Beta, and Gamma variants as well as T helper (Th)1 CD4+-biased T cell responses. Although anti-receptor-binding domain (RBD)-specific antibody responses are initially predominant, the third immunization boosts significant non-RBD antibody titers. Challenging vaccinated animals with SARS-CoV-2 shows a complete protection through sterilizing immunity, which correlates with the presence of nasopharyngeal anti-S immunoglobulin G (IgG) and IgA titers. Thus, the S-LV approach is an efficient and safe vaccine candidate based on a proven classical approach for further development and clinical testing.


Assuntos
Vacinas contra COVID-19/administração & dosagem , COVID-19/prevenção & controle , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinação/métodos , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , COVID-19/epidemiologia , COVID-19/imunologia , COVID-19/virologia , Vacinas contra COVID-19/imunologia , Chlorocebus aethiops , Modelos Animais de Doenças , Células HEK293 , Humanos , Imunoglobulina A/imunologia , Imunoglobulina G/imunologia , Lipossomos , Macaca fascicularis , Masculino , Pandemias/prevenção & controle , Células Th1/imunologia , Resultado do Tratamento , Vacinas de Partículas Semelhantes a Vírus/imunologia , Células Vero
4.
Elife ; 102021 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-33871352

RESUMO

The HIV-1 gp120/gp41 trimer undergoes a series of conformational changes in order to catalyze gp41-induced fusion of viral and cellular membranes. Here, we present the crystal structure of gp41 locked in a fusion intermediate state by an MPER-specific neutralizing antibody. The structure illustrates the conformational plasticity of the six membrane anchors arranged asymmetrically with the fusion peptides and the transmembrane regions pointing into different directions. Hinge regions located adjacent to the fusion peptide and the transmembrane region facilitate the conformational flexibility that allows high-affinity binding of broadly neutralizing anti-MPER antibodies. Molecular dynamics simulation of the MPER Ab-stabilized gp41 conformation reveals a possible transition pathway into the final post-fusion conformation with the central fusion peptides forming a hydrophobic core with flanking transmembrane regions. This suggests that MPER-specific broadly neutralizing antibodies can block final steps of refolding of the fusion peptide and the transmembrane region, which is required for completing membrane fusion.


Assuntos
Anticorpos Amplamente Neutralizantes/metabolismo , Anticorpos Anti-HIV/metabolismo , Proteína gp41 do Envelope de HIV/antagonistas & inibidores , HIV-1/imunologia , Anticorpos de Domínio Único/metabolismo , Especificidade de Anticorpos , Sítios de Ligação de Anticorpos , Anticorpos Amplamente Neutralizantes/imunologia , Células HEK293 , Anticorpos Anti-HIV/imunologia , Proteína gp41 do Envelope de HIV/imunologia , Proteína gp41 do Envelope de HIV/metabolismo , Humanos , Bicamadas Lipídicas , Fusão de Membrana , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Estabilidade Proteica , Anticorpos de Domínio Único/imunologia , Relação Estrutura-Atividade
5.
Viruses ; 12(11)2020 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-33114242

RESUMO

HIV-1 vaccine research has obtained an enormous boost since the discovery of many broadly neutralizing antibodies (bnAbs) targeting all accessible sites on the HIV-1 envelope glycoprotein (Env). This in turn facilitated high-resolution structures of the Env glycoprotein in complex with bnAbs. Here we focus on gp41, its highly conserved heptad repeat region 1 (HR1), the fusion peptide (FP) and the membrane-proximal external region (MPER). Notably, the broadest neutralizing antibodies target MPER. Both gp41 HR1 and MPER are only fully accessible once receptor-induced conformational changes have taken place, although some studies suggest access to MPER in the close to native Env conformation. We summarize the data on the structure and function of neutralizing antibodies targeting gp41 HR1, FP and MPER and we review their access to Env and their complex formation with gp41 HR1, MPER peptides and FP within native Env. We further discuss MPER bnAb binding to lipids and the role of somatic mutations in recognizing a bipartite epitope composed of the conserved MPER sequence and membrane components. The problematic of gp41 HR1 access and MPER bnAb auto- and polyreactivity is developed in the light of inducing such antibodies by vaccination.


Assuntos
Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/imunologia , Anticorpos Anti-HIV/imunologia , Proteína gp41 do Envelope de HIV/imunologia , Vacinas contra a AIDS/imunologia , Animais , Epitopos/imunologia , Proteína gp41 do Envelope de HIV/genética , HIV-1/genética , HIV-1/imunologia , Humanos , Camundongos , Mutação
6.
Nucleic Acids Res ; 45(6): 3353-3368, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28126917

RESUMO

Influenza polymerase replicates, via a complementary RNA intermediate (cRNA), and transcribes the eight viral RNA (vRNA) genome segments. To initiate RNA synthesis it is bound to the conserved 5΄ and 3΄ extremities of the vRNA or cRNA (the 'promoter'). 5΄-3΄ base-pairing in the distal promoter region is essential to position the template RNA at the polymerase active site, as shown by a new crystal structure with the 3΄ end threading through the template entry tunnel. We develop fluorescence polarization assays to quantify initiation of cap-primed (transcription) or unprimed (replication) RNA synthesis by recombinant influenza B polymerase bound to the vRNA or cRNA promoter. The rate-limiting step is formation of a primed initiation complex with minimally ApG required to stabilize the 3΄ end of the template within the active-site. Polymerase bound to the vRNA promoter initiates RNA synthesis terminally, while the cRNA promoter directs internal initiation at a significantly lower rate. Progression to elongation requires breaking the promoter 5΄-3΄ base-pairing region and favourable compensation by the emerging template-product base-pairs. The RNA synthesis assay is adaptable to high-throughput screening for polymerase inhibitors. In a pilot study, we find that initiation at the cRNA promoter is unusually susceptible to inhibition by 2΄F-2΄dNTPs.


Assuntos
RNA Polimerases Dirigidas por DNA/metabolismo , Vírus da Influenza B/enzimologia , RNA Viral/biossíntese , Proteínas Virais/metabolismo , Pareamento de Bases , RNA Polimerases Dirigidas por DNA/antagonistas & inibidores , RNA Polimerases Dirigidas por DNA/química , Polarização de Fluorescência , Vírus da Influenza B/genética , Vírus da Influenza B/fisiologia , Regiões Promotoras Genéticas , RNA Viral/química , Transcrição Gênica , Proteínas Virais/química , Replicação Viral
7.
Mol Cell ; 61(1): 125-37, 2016 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-26711008

RESUMO

Influenza virus polymerase transcribes or replicates the segmented RNA genome (vRNA) into respectively viral mRNA or full-length copies and initiates RNA synthesis by binding the conserved 3' and 5' vRNA ends (the promoter). In recent structures of promoter-bound polymerase, the cap-binding and endonuclease domains are configured for cap snatching, which generates capped transcription primers. Here, we present a FluB polymerase structure with a bound complementary cRNA 5' end that exhibits a major rearrangement of the subdomains within the C-terminal two-thirds of PB2 (PB2-C). Notably, the PB2 nuclear localization signal (NLS)-containing domain translocates ∼90 Što bind to the endonuclease domain. FluA PB2-C alone and RNA-free FluC polymerase are similarly arranged. Biophysical and cap-dependent endonuclease assays show that in solution the polymerase explores different conformational distributions depending on which RNA is bound. The inherent flexibility of the polymerase allows it to adopt alternative conformations that are likely important during polymerase maturation into active progeny RNPs.


Assuntos
Gammainfluenzavirus/enzimologia , Virus da Influenza A Subtipo H5N1/enzimologia , Vírus da Influenza B/enzimologia , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Cristalografia por Raios X , Humanos , Virus da Influenza A Subtipo H5N1/genética , Vírus da Influenza B/genética , Gammainfluenzavirus/genética , Lasers , Espectrometria de Massas , Modelos Moleculares , Dados de Sequência Molecular , Sinais de Localização Nuclear/metabolismo , Domínios e Motivos de Interação entre Proteínas , RNA Viral/metabolismo , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/genética , Ribonucleoproteínas/metabolismo , Espalhamento a Baixo Ângulo , Relação Estrutura-Atividade , Proteínas Virais/química , Proteínas Virais/genética
8.
Nature ; 516(7531): 355-60, 2014 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-25409142

RESUMO

The influenza virus polymerase transcribes or replicates the segmented RNA genome (viral RNA) into viral messenger RNA or full-length copies. To initiate RNA synthesis, the polymerase binds to the conserved 3' and 5' extremities of the viral RNA. Here we present the crystal structure of the heterotrimeric bat influenza A polymerase, comprising subunits PA, PB1 and PB2, bound to its viral RNA promoter. PB1 contains a canonical RNA polymerase fold that is stabilized by large interfaces with PA and PB2. The PA endonuclease and the PB2 cap-binding domain, involved in transcription by cap-snatching, form protrusions facing each other across a solvent channel. The 5' extremity of the promoter folds into a compact hook that is bound in a pocket formed by PB1 and PA close to the polymerase active site. This structure lays the basis for an atomic-level mechanistic understanding of the many functions of influenza polymerase, and opens new opportunities for anti-influenza drug design.


Assuntos
RNA Polimerases Dirigidas por DNA/química , Vírus da Influenza A/enzimologia , RNA Viral/química , Sítios de Ligação , Cristalização , Modelos Moleculares , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , Subunidades Proteicas/química
9.
Nature ; 516(7531): 361-6, 2014 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-25409151

RESUMO

Influenza virus polymerase uses a capped primer, derived by 'cap-snatching' from host pre-messenger RNA, to transcribe its RNA genome into mRNA and a stuttering mechanism to generate the poly(A) tail. By contrast, genome replication is unprimed and generates exact full-length copies of the template. Here we use crystal structures of bat influenza A and human influenza B polymerases (FluA and FluB), bound to the viral RNA promoter, to give mechanistic insight into these distinct processes. In the FluA structure, a loop analogous to the priming loop of flavivirus polymerases suggests that influenza could initiate unprimed template replication by a similar mechanism. Comparing the FluA and FluB structures suggests that cap-snatching involves in situ rotation of the PB2 cap-binding domain to direct the capped primer first towards the endonuclease and then into the polymerase active site. The polymerase probably undergoes considerable conformational changes to convert the observed pre-initiation state into the active initiation and elongation states.


Assuntos
RNA Polimerases Dirigidas por DNA/metabolismo , Vírus da Influenza A/enzimologia , Vírus da Influenza B/enzimologia , Modelos Moleculares , Capuzes de RNA , RNA Viral/biossíntese , RNA Viral/química , Domínio Catalítico , Cristalização , RNA Polimerases Dirigidas por DNA/química , Regulação Viral da Expressão Gênica , Vírus da Influenza A/química , Vírus da Influenza B/química , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , Capuzes de RNA/química , Capuzes de RNA/metabolismo , Replicação Viral
10.
PLoS One ; 9(1): e84973, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24454773

RESUMO

Orthomyxovirus Influenza A virus (IAV) heterotrimeric polymerase performs transcription of viral mRNAs by cap-snatching, which involves generation of capped primers by host pre-mRNA binding via the PB2 subunit cap-binding site and cleavage 10-13 nucleotides from the 5' cap by the PA subunit endonuclease. Thogotoviruses, tick-borne orthomyxoviruses that includes Thogoto (THOV), Dhori (DHOV) and Jos (JOSV) viruses, are thought to perform cap-snatching by cleaving directly after the cap and thus have no heterogeneous, host-derived sequences at the 5' extremity of their mRNAs. Based on recent work identifying the cap-binding and endonuclease domains in IAV polymerase, we determined the crystal structures of two THOV PB2 domains, the putative cap-binding and the so-called '627-domain', and the structures of the putative endonuclease domains (PA-Nter) of THOV and DHOV. Despite low sequence similarity, corresponding domains have the same fold confirming the overall architectural similarity of orthomyxovirus polymerases. However the putative Thogotovirus cap-snatching domains in PA and PB2 have non-conservative substitutions of key active site residues. Biochemical analysis confirms that, unlike the IAV domains, the THOV and DHOV PA-Nter domains do not bind divalent cations and have no endonuclease activity and the THOV central PB2 domain does not bind cap analogues. On the other hand, sequence analysis suggests that other, non-influenza, orthomyxoviruses, such as salmon anemia virus (isavirus) and Quaranfil virus likely conserve active cap-snatching domains correlating with the reported occurrence of heterogeneous, host-derived sequences at the 5' end of the mRNAs of these viruses. These results highlight the unusual nature of transcription initiation by Thogotoviruses.


Assuntos
RNA Polimerases Dirigidas por DNA/metabolismo , Orthomyxoviridae/genética , Capuzes de RNA , Sequência de Aminoácidos , RNA Polimerases Dirigidas por DNA/química , Dados de Sequência Molecular , Orthomyxoviridae/enzimologia , Conformação Proteica , Homologia de Sequência de Aminoácidos
11.
J Med Chem ; 56(21): 8915-30, 2013 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-24134208

RESUMO

The heterotrimeric influenza virus polymerase performs replication and transcription of viral RNA in the nucleus of infected cells. Transcription by "cap-snatching" requires that host-cell pre-mRNAs are bound via their 5' cap to the PB2 subunit. Thus, the PB2 cap-binding site is potentially a good target for new antiviral drugs that will directly inhibit viral replication. Docking studies using the structure of the PB2 cap-binding domain suggested that 7-alkylguanine derivatives substituted at position N-9 and N-2 could be good candidates. Four series of 7,9-di- and 2,7,9-trialkyl guanine derivatives were synthesized and evaluated by an AlphaScreen assay in competition with a biotinylated cap analogue. Three synthesized compounds display potent in vitro activity with IC50 values lower than 10 µM. High-resolution X-ray structures of three inhibitors in complex with the H5N1 PB2 cap-binding domain confirmed the binding mode and provide detailed information for further compound optimization.


Assuntos
Guanina/análogos & derivados , Vírus da Influenza A/enzimologia , RNA Polimerase Dependente de RNA/antagonistas & inibidores , Proteínas Virais/antagonistas & inibidores , Sítios de Ligação/efeitos dos fármacos , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Guanina/síntese química , Guanina/química , Guanina/farmacologia , Modelos Moleculares , Estrutura Molecular , RNA Polimerase Dependente de RNA/metabolismo , Relação Estrutura-Atividade , Proteínas Virais/metabolismo
12.
Nat Commun ; 4: 1429, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23385574

RESUMO

The typical bullet shape of Rhabdoviruses is thought to rely on the matrix protein for stabilizing the nucleocapsid coil. Here we scrutinize the morphology of purified and recombinant nucleocapsids of vesicular stomatitis virus in vitro. We elucidate pH and ionic strength conditions for their folding into conical tips and further growth into whole bullets, and provide cryo-electron microscopy reconstructions of the bullet tip and the helical trunk. We address conformational variability of the reconstituted nucleocapsids and the issue of constraints imposed by the binding of matrix protein. Our findings bridge the gap between the isolated nucleoprotein-RNA string in its form of an undulating ribbon, and the tight bullet-shaped virion skeleton.


Assuntos
Nucleocapsídeo/ultraestrutura , Vírus da Estomatite Vesicular Indiana/ultraestrutura , Microscopia Crioeletrônica , Conformação de Ácido Nucleico , Nucleoproteínas/metabolismo , RNA Viral/ultraestrutura , Proteínas da Matriz Viral/metabolismo
13.
Virologie (Montrouge) ; 14(6): 435-445, 2010 Dec 01.
Artigo em Francês | MEDLINE | ID: mdl-36151627

RESUMO

Influenza virus polymerase transcribes and replicates the viral RNA genome within the context of a ribonucleoprotein complex that has been hitherto remarkably intractable to structural analysis. In the last three years, crystal structures of independent domains covering roughly half of the heterotrimeric polymerase have been determined. These include the cap-binding and endonuclease domains, critical for the unique cap-snatching mechanism of mRNA transcription, and the major inter-subunit interfaces. In addition a cryoelectron microscopy structure of the entire ribonucleoprotein complex has been determined opening the way to the construction of a quasi-atomic model of the influenza replication machinery. These results provide the first detailed structure-function insights into polymerase assembly, transcription and host adaptation and will have an impact on anti-influenza drug design.

14.
J Med Chem ; 52(12): 3666-78, 2009 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-19456100

RESUMO

The adenovirus serotype Ad37 binds to and infects human corneal epithelial (HCE) cells through attachment to cellular glycoproteins carrying terminal sialic acids. By use of the crystallographic structure of the sialic acid-interacting domain of the Ad37 fiber protein in complex with sialyllactose, a set of N-acyl modified sialic acids were designed to improve binding affinity through increased hydrophobic interactions. These N-acyl modified sialic acids and their corresponding multivalent human serum albumin (HSA) conjugates were synthesized and tested in Ad37 cell binding and cell infectivity assays. Compounds bearing small substituents were as effective inhibitors as sialic acid. X-ray crystallography and overlays with the Ad37-sialyllactose complex showed that the N-acyl modified sialic acids were positioned in the same orientation as sialic acid. Their multivalent counterparts achieved a strong multivalency effect and were more effective to prevent infection than the monomers. Unfortunately, they were less active as inhibitors than multivalent sialic acid.


Assuntos
Adenovírus Humanos/efeitos dos fármacos , Desenho de Fármacos , Ceratoconjuntivite/epidemiologia , Ceratoconjuntivite/virologia , Ácidos Siálicos/síntese química , Ácidos Siálicos/farmacologia , Sítios de Ligação , Simulação por Computador , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Humanos , Modelos Químicos , Modelos Moleculares , Conformação Molecular , Albumina Sérica/química , Ácidos Siálicos/química , Relação Estrutura-Atividade
15.
PLoS Pathog ; 4(8): e1000136, 2008 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-18769709

RESUMO

Understanding how avian influenza viruses adapt to human hosts is critical for the monitoring and prevention of future pandemics. Host specificity is determined by multiple sites in different viral proteins, and mutation of only a limited number of these sites can lead to inter-species transmission. Several of these sites have been identified in the viral polymerase, the best characterised being position 627 in the PB2 subunit. Efficient viral replication at the relatively low temperature of the human respiratory tract requires lysine 627 rather than the glutamic acid variant found systematically in avian viruses. However, the molecular mechanism by which any of these host specific sites determine host range are unknown, although adaptation to host factors is frequently evoked. We used ESPRIT, a library screening method, to identify a new PB2 domain that contains a high density of putative host specific sites, including residue 627. The X-ray structure of this domain (denoted the 627-domain) exhibits a novel fold with the side-chain of Lys627 solvent exposed. The structure of the K627E mutated domain shows no structural differences but the charge reversal disrupts a striking basic patch on the domain surface. Five other recently proposed host determining sites of PB2 are also located on the 627-domain surface. The structure of the complete C-terminal region of PB2 comprising the 627-domain and the previously identified NLS-domain, which binds the host nuclear import factor importin alpha, was also determined. The two domains are found to pack together with a largely hydrophilic interface. These data enable a three-dimensional mapping of approximately half of PB2 sites implicated in cross-species transfer onto a single structural unit. Their surface location is consistent with roles in interactions with other viral proteins or host factors. The identification and structural characterization of these well-defined PB2 domains will help design experiments to elucidate the effects of mutations on polymerase-host factor interactions.


Assuntos
Adaptação Fisiológica , Substituição de Aminoácidos , Vírus da Influenza A Subtipo H3N2/enzimologia , Mutação de Sentido Incorreto , RNA Polimerase Dependente de RNA/química , Proteínas Virais/química , Cristalografia por Raios X , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A Subtipo H3N2/genética , Influenza Humana/enzimologia , Influenza Humana/genética , Influenza Humana/metabolismo , Estrutura Terciária de Proteína/genética , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , alfa Carioferinas/química , alfa Carioferinas/metabolismo
16.
Nat Struct Mol Biol ; 15(5): 500-6, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18454157

RESUMO

Influenza virus mRNAs are synthesized by the trimeric viral polymerase using short capped primers obtained by a 'cap-snatching' mechanism. The polymerase PB2 subunit binds the 5' cap of host pre-mRNAs, which are cleaved after 10-13 nucleotides by the PB1 subunit. Using a library-screening method, we identified an independently folded domain of PB2 that has specific cap binding activity. The X-ray structure of the domain with bound cap analog m(7)GTP at 2.3-A resolution reveals a previously unknown fold and a mode of ligand binding that is similar to, but distinct from, other cap binding proteins. Binding and functional studies with point mutants confirm that the identified site is essential for cap binding in vitro and cap-dependent transcription in vivo by the trimeric polymerase complex. These findings clarify the nature of the cap binding site in PB2 and will allow efficient structure-based design of new anti-influenza compounds inhibiting viral transcription.


Assuntos
Vírus da Influenza A/metabolismo , Capuzes de RNA/metabolismo , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo , Linhagem Celular , Humanos , Vírus da Influenza A/química , Mutação Puntual , Estrutura Terciária de Proteína , Análogos de Capuz de RNA/metabolismo , RNA Polimerase Dependente de RNA/genética , Transcrição Gênica , Proteínas Virais/genética
17.
Nat Struct Mol Biol ; 14(3): 229-33, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17310249

RESUMO

The trimeric influenza virus polymerase, comprising subunits PA, PB1 and PB2, is responsible for transcription and replication of the segmented viral RNA genome. Using a novel library-based screening technique called expression of soluble proteins by random incremental truncation (ESPRIT), we identified an independently folded C-terminal domain from PB2 and determined its solution structure by NMR. Using green fluorescent protein fusions, we show that both the domain and the full-length PB2 subunit are efficiently imported into the nucleus dependent on a previously overlooked bipartite nuclear localization sequence (NLS). The crystal structure of the domain complexed with human importin alpha5 shows how the last 20 residues unfold to permit binding to the import factor. The domain contains three surface residues implicated in adaptation from avian to mammalian hosts. One of these tethers the NLS-containing peptide to the core of the domain in the unbound state.


Assuntos
Transporte Ativo do Núcleo Celular , Núcleo Celular/metabolismo , Orthomyxoviridae/enzimologia , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Sobrevivência Celular , Cristalografia por Raios X , Humanos , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Sinais de Localização Nuclear , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Solubilidade , Soluções , alfa Carioferinas/química
18.
RNA ; 12(10): 1817-24, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16931876

RESUMO

UPF1 is an essential eukaryotic RNA helicase that plays a key role in various mRNA degradation pathways, notably nonsense-mediated mRNA decay (NMD). In combination with UPF2 and UPF3, it forms part of the surveillance complex that detects mRNAs containing premature stop codons and triggers their degradation in all organisms studied from yeast to human. We describe the 3 A resolution crystal structure of the highly conserved cysteine-histidine-rich domain of human UPF1 and show that it is a unique combination of three zinc-binding motifs arranged into two tandem modules related to the RING-box and U-box domains of ubiquitin ligases. This UPF1 domain interacts with UPF2, and we identified by mutational analysis residues in two distinct conserved surface regions of UPF1 that mediate this interaction. UPF1 residues we identify as important for the interaction with UPF2 are not conserved in UPF1 homologs from certain unicellular parasites that also appear to lack UPF2 in their genomes.


Assuntos
RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transativadores/química , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Códon sem Sentido , Cristalografia por Raios X , Humanos , Técnicas In Vitro , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína , RNA Helicases , Proteínas de Ligação a RNA , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Transativadores/genética , Zinco/química
19.
J Virol ; 78(14): 7727-36, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15220447

RESUMO

Adenovirus serotype 37 (Ad37) belongs to species D and can cause epidemic keratoconjunctivitis, whereas the closely related Ad19p does not. Primary cell attachment by adenoviruses is mediated through receptor binding of the knob domain of the fiber protein. The knobs of Ad37 and Ad19p differ at only two positions, Lys240Glu and Asn340Asp. We report the high-resolution crystal structures of the Ad37 and Ad19p knobs, both native and in complex with sialic acid, which has been proposed as a receptor for Ad37. Overall, the Ad37 and Ad19p knobs are very similar to previously reported knob structures, especially to that of Ad5, which binds the coxsackievirus-adenovirus receptor (CAR). Ad37 and Ad19p knobs are structurally identical with the exception of the changed side chains and are structurally most similar to CAR-binding knobs (e.g., that of Ad5) rather than non-CAR-binding knobs (e.g., that of Ad3). The two mutations in Ad19p result in a partial loss of the exceptionally high positive surface charge of the Ad37 knob but do not affect sialic acid binding. This site is located on the top of the trimer and binds both alpha(2,3) and alpha(2,6)-linked sialyl-lactose, although only the sialic acid residue makes direct contact. Amino acid alignment suggests that the sialic acid binding site is conserved in several species D serotypes. Our results show that the altered viral tropism and cell binding of Ad19p relative to those of Ad37 are not explained by a different binding ability toward sialyl-lactose.


Assuntos
Adenovírus Humanos/metabolismo , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Adenovírus Humanos/patogenicidade , Sequência de Aminoácidos , Sítios de Ligação , Proteínas do Capsídeo/genética , Linhagem Celular , Cristalização , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Receptores Virais/química , Receptores Virais/genética , Receptores Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...