Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 3: 2871, 2013 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-24129600

RESUMO

The influenza virus neuraminidase (NA) is essential for the virus life cycle. The rise of resistance mutations against current antiviral therapies has increased the need for the development of novel inhibitors. Recent efforts have targeted a cavity adjacent to the catalytic site (the 150-cavity) in addition to the primary catalytic subsite in order to increase specificity and reduce the likelihood of resistance. This study details structural and in vitro analyses of a class of inhibitors that bind uniquely in both subsites. Crystal structures of three inhibitors show occupation of the 150-cavity in two distinct and novel binding modes. We believe these are the first nanomolar inhibitors of NA to be characterized in this way. Furthermore, we show that one inhibitor, binding within the catalytic site, offers reduced susceptibility to known resistance mutations via increased flexibility of a pendant pentyloxy group and the ability to pivot about a strong hydrogen-bonding network.


Assuntos
Antivirais/química , Inibidores Enzimáticos/química , Vírus da Influenza A/enzimologia , Neuraminidase/química , Proteínas Virais/química , Animais , Antivirais/metabolismo , Antivirais/farmacologia , Domínio Catalítico , Linhagem Celular , Farmacorresistência Viral , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/enzimologia , Vírus da Influenza A/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Conformação Molecular , Estrutura Molecular , Neuraminidase/antagonistas & inibidores , Neuraminidase/metabolismo , Oseltamivir/química , Oseltamivir/farmacologia , Ligação Proteica , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo
2.
J Med Chem ; 55(20): 8963-8, 2012 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-23017008

RESUMO

A series of C3 O-functionalized 2-acetamido-2-deoxy-Δ4-ß-D-glucuronides were synthesized to explore noncharge interactions in subsite 2 of the influenza virus sialidase active site. In complex with A/N8 sialidase, the parent compound (C3 OH) inverts its solution conformation to bind with all substituents well positioned in the active site. The parent compound inhibits influenza virus sialidase at a sub-µM level; the introduction of small alkyl substituents or an acetyl group at C3 is also tolerated.


Assuntos
Acetamidas/química , Antivirais/química , Glucuronídeos/química , Neuraminidase/química , Orthomyxoviridae/enzimologia , Acetamidas/síntese química , Domínio Catalítico , Ensaios Enzimáticos , Fluorometria , Glucuronídeos/síntese química , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Neuraminidase/antagonistas & inibidores , Eletricidade Estática , Relação Estrutura-Atividade
3.
J Virol ; 86(23): 13095-8, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22993153

RESUMO

Conserved tryptophan-187 facilitates homodimerization of the influenza A virus NS1 protein effector domain. We generated a mutant influenza virus strain expressing NS1-W187R to destabilize this self-interaction. NS1-W187R protein exhibited lower double-stranded RNA (dsRNA)-binding activity, showed a temporal redistribution during infection, and was minimally compromised for interferon antagonism. The mutant virus replicated similarly to the wild type in vitro, but it was slightly attenuated for replication in mice, causing notably reduced morbidity and mortality. These data suggest biological relevance for the W187-mediated homotypic interaction of NS1.


Assuntos
Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/patogenicidade , Modelos Moleculares , Proteínas não Estruturais Virais/química , Replicação Viral/fisiologia , Animais , Western Blotting , Dimerização , Cães , Técnica Indireta de Fluorescência para Anticorpo , Vírus da Influenza A Subtipo H1N1/fisiologia , Luciferases , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos C57BL , Mutação de Sentido Incorreto/genética , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
4.
Org Biomol Chem ; 10(43): 8628-39, 2012 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-22976385

RESUMO

Novel 3-C-alkylated-Neu5Ac2en derivatives have been designed to target the expanded active site cavity of influenza virus sialidases with an open 150-loop, currently seen in X-ray crystal structures of influenza A virus group-1 (N1, N4, N5, N8), but not group-2 (N2, N9), sialidases. The compounds show selectivity for inhibition of H5N1 and pdm09 H1N1 sialidases over an N2 sialidase, providing evidence of the relative 150-loop flexibility of these sialidases. In a complex with N8 sialidase, the C3 substituent of 3-phenylally-Neu5Ac2en occupies the 150-cavity while the central ring and the remaining substituents bind the active site as seen for the unsubstituted template. This new class of inhibitors, which can 'trap' the open 150-loop form of the sialidase, should prove useful as probes of 150-loop flexibility.


Assuntos
Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Vírus da Influenza A/enzimologia , Ácido N-Acetilneuramínico/análogos & derivados , Neuraminidase/antagonistas & inibidores , Alquilação , Domínio Catalítico/efeitos dos fármacos , Cristalografia por Raios X , Desenho de Fármacos , Inibidores Enzimáticos/química , Modelos Moleculares , Estrutura Molecular , Ácido N-Acetilneuramínico/síntese química , Ácido N-Acetilneuramínico/química , Ácido N-Acetilneuramínico/farmacologia , Neuraminidase/metabolismo , Maleabilidade/efeitos dos fármacos , Relação Estrutura-Atividade
5.
Artigo em Inglês | MEDLINE | ID: mdl-21821881

RESUMO

The effector domain (ED) of the influenza virus virulence factor NS1 is capable of interaction with a variety of cellular and viral targets, although regulation of these events is poorly understood. Introduction of a W187A mutation into the ED abolishes dimer formation; however, strand-strand interactions between mutant NS1 ED monomers have been observed in two previous crystal forms. A new condition for crystallization of this protein [0.1 M Bis-Tris pH 6.0, 0.2 M NaCl, 22%(w/v) PEG 3350, 20 mM xylitol] was discovered using the hanging-drop vapour-diffusion method. Diffraction data extending to 1.8 Šresolution were collected from a crystal grown in the presence of 40 mM thieno[2,3-b]pyridin-2-ylmethanol. It was observed that there is conservation of the strand-strand interface in crystals of this monomeric NS1 ED in three different space groups. This observation, coupled with conformational changes in the interface region, suggests a potential role for ß-sheet augmentation in NS1 function.


Assuntos
Orthomyxoviridae/química , Proteínas não Estruturais Virais/química , Cristalografia por Raios X , Modelos Moleculares , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
6.
PLoS One ; 6(3): e17946, 2011 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-21464929

RESUMO

Influenza A virus NS1 protein is a multifunctional virulence factor consisting of an RNA binding domain (RBD), a short linker, an effector domain (ED), and a C-terminal 'tail'. Although poorly understood, NS1 multimerization may autoregulate its actions. While RBD dimerization seems functionally conserved, two possible apo ED dimers have been proposed (helix-helix and strand-strand). Here, we analyze all available RBD, ED, and full-length NS1 structures, including four novel crystal structures obtained using EDs from divergent human and avian viruses, as well as two forms of a monomeric ED mutant. The data reveal the helix-helix interface as the only strictly conserved ED homodimeric contact. Furthermore, a mutant NS1 unable to form the helix-helix dimer is compromised in its ability to bind dsRNA efficiently, implying that ED multimerization influences RBD activity. Our bioinformatical work also suggests that the helix-helix interface is variable and transient, thereby allowing two ED monomers to twist relative to one another and possibly separate. In this regard, we found a mAb that recognizes NS1 via a residue completely buried within the ED helix-helix interface, and which may help highlight potential different conformational populations of NS1 (putatively termed 'helix-closed' and 'helix-open') in virus-infected cells. 'Helix-closed' conformations appear to enhance dsRNA binding, and 'helix-open' conformations allow otherwise inaccessible interactions with host factors. Our data support a new model of NS1 regulation in which the RBD remains dimeric throughout infection, while the ED switches between several quaternary states in order to expand its functional space. Such a concept may be applicable to other small multifunctional proteins.


Assuntos
Vírus da Influenza A/metabolismo , Modelos Biológicos , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Animais , Linhagem Celular , Biologia Computacional , Sequência Conservada , Cristalografia por Raios X , Cães , Humanos , Interferon beta/genética , Proteínas Mutantes/química , Mutação/genética , Maleabilidade , Ligação Proteica , Multimerização Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , RNA Viral/metabolismo , Proteínas não Estruturais Virais/antagonistas & inibidores
7.
Virology ; 413(2): 169-82, 2011 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-21353280

RESUMO

We have utilized glycan microarray technology to determine the receptor binding properties of early isolates from the recent 2009 H1N1 human pandemic (pdmH1N1), and compared them to North American swine influenza isolates from the same year, as well as past seasonal H1N1 human isolates. We showed that the pdmH1N1 strains, as well as the swine influenza isolates examined, bound almost exclusively to glycans with α2,6-linked sialic acid with little binding detected for α2,3-linked species. This is highlighted by pair-wise comparisons between compounds with identical glycan backbones, differing only in the chemistry of their terminal linkages. The overall similarities in receptor binding profiles displayed by pdmH1N1 strains and swine isolates indicate that little or no adaptation appeared to be necessary in the binding component of HA for transmission from pig to human, and subsequent human to human spread.


Assuntos
Vírus da Influenza A Subtipo H1N1/fisiologia , Influenza Humana/virologia , Infecções por Orthomyxoviridae/veterinária , Receptores Virais/metabolismo , Doenças dos Suínos/virologia , Ligação Viral , Aglutinação , Animais , Sítios de Ligação , Eritrócitos , Hemaglutininas/química , Hemaglutininas/genética , Hemaglutininas/metabolismo , Humanos , Influenza Humana/epidemiologia , Análise em Microsséries/métodos , Modelos Moleculares , Infecções por Orthomyxoviridae/epidemiologia , Infecções por Orthomyxoviridae/virologia , Pandemias , Filogenia , Ligação Proteica , Conformação Proteica , Estações do Ano , Suínos , Doenças dos Suínos/epidemiologia
8.
Antimicrob Agents Chemother ; 55(6): 2942-52, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21422222

RESUMO

Neuraminidase (NA) inhibitors (NIs) are the first line of defense against influenza virus. Reverse genetics experiments allow the study of resistance mechanisms by anticipating the impacts of mutations to the virus. To look at the possibility of an increased effect on the resistance phenotype of a combination of framework mutations, known to confer resistance to oseltamivir or zanamivir, with limited effect on virus fitness, we constructed 4 viruses by reverse genetics in the A/Moscow/10/99 H3N2 background containing double mutations in their neuraminidase genes: E119D+I222L, E119V+I222L, D198N+I222L, and H274Y+I222L (N2 numbering). Among the viruses produced, the E119D+I222L mutant virus was not able to grow without bacterial NA complementation and the D198N+I222L mutant and H274Y+I222L mutant were not stable after passages in MDCK cells. The E119V+I222L mutant was stable after five passages in MDCK cells. This E119V-and-I222L combination had a combinatorial effect on oseltamivir resistance. The total NA activity of the E119V+I222L mutant was low (5% compared to that of the wild-type virus). This drop in NA activity resulted from a decreased NA quantity in the virion in comparison to that of the wild-type virus (1.4% of that of the wild type). In MDCK-SIAT1 cells, the E119V+I222L mutant virus did not present a replicative advantage over the wild-type virus, even in the presence of oseltamivir. Double mutations combining two framework mutations in the NA gene still have to be monitored, as they could induce a high level of resistance to NIs, without impairing the NA affinity. Our study allows a better understanding of the diversity of the mechanisms of resistance to NIs.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A Subtipo H3N2/efeitos dos fármacos , Mutação , Neuraminidase/genética , Oseltamivir/farmacologia , Animais , Sítios de Ligação , Células Cultivadas , Cães , Farmacorresistência Viral , Vírus da Influenza A Subtipo H3N2/enzimologia , Vírus da Influenza A Subtipo H3N2/genética , Vírus da Influenza A Subtipo H3N2/crescimento & desenvolvimento , Neuraminidase/antagonistas & inibidores , Neuraminidase/metabolismo , Vírion/enzimologia
9.
Nat Commun ; 1: 113, 2010 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-21081911

RESUMO

Influenza virus sialidase has an essential role in the virus' life cycle. Two distinct groups of influenza A virus sialidases have been established, that differ in the flexibility of the '150-loop', providing a more open active site in the apo form of the group-1 compared to group-2 enzymes. In this study we show, through a multidisciplinary approach, that novel sialic acid-based derivatives can exploit this structural difference and selectively inhibit the activity of group-1 sialidases. We also demonstrate that group-1 sialidases from drug-resistant mutant influenza viruses are sensitive to these designed compounds. Moreover, we have determined, by protein X-ray crystallography, that these inhibitors lock open the group-1 sialidase flexible 150-loop, in agreement with our molecular modelling prediction. This is the first direct proof that compounds may be developed to selectively target the pandemic A/H1N1, avian A/H5N1 and other group-1 sialidase-containing viruses, based on an open 150-loop conformation of the enzyme.

10.
Proc Natl Acad Sci U S A ; 107(5): 1954-9, 2010 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-20133840

RESUMO

Seasonal epidemics and periodic worldwide pandemics caused by influenza A viruses are of continuous concern. The viral nonstructural (NS1) protein is a multifunctional virulence factor that antagonizes several host innate immune defenses during infection. NS1 also directly stimulates class IA phosphoinositide 3-kinase (PI3K) signaling, an essential cell survival pathway commonly mutated in human cancers. Here, we present a 2.3-A resolution crystal structure of the NS1 effector domain in complex with the inter-SH2 (coiled-coil) domain of p85beta, a regulatory subunit of PI3K. Our data emphasize the remarkable isoform specificity of this interaction, and provide insights into the mechanism by which NS1 activates the PI3K (p85beta:p110) holoenzyme. A model of the NS1:PI3K heterotrimeric complex reveals that NS1 uses the coiled-coil as a structural tether to sterically prevent normal inhibitory contacts between the N-terminal SH2 domain of p85beta and the p110 catalytic subunit. Furthermore, in this model, NS1 makes extensive contacts with the C2/kinase domains of p110, and a small acidic alpha-helix of NS1 sits adjacent to the highly basic activation loop of the enzyme. During infection, a recombinant influenza A virus expressing NS1 with charge-disruption mutations in this acidic alpha-helix is unable to stimulate the production of phosphatidylinositol 3,4,5-trisphosphate or the phosphorylation of Akt. Despite this, the charge-disruption mutations in NS1 do not affect its ability to interact with the p85beta inter-SH2 domain in vitro. Overall, these data suggest that both direct binding of NS1 to p85beta (resulting in repositioning of the N-terminal SH2 domain) and possible NS1:p110 contacts contribute to PI3K activation.


Assuntos
Vírus da Influenza A Subtipo H1N1/metabolismo , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Animais , Sequência de Bases , Domínio Catalítico , Bovinos , Linhagem Celular , Cristalografia por Raios X , Primers do DNA/genética , Cães , Ativação Enzimática , Humanos , Técnicas In Vitro , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/patogenicidade , Modelos Moleculares , Complexos Multiproteicos , Mutagênese Sítio-Dirigida , Domínios e Motivos de Interação entre Proteínas , Proteínas não Estruturais Virais/genética , Domínios de Homologia de src
11.
Virology ; 396(1): 94-105, 2010 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-19880155

RESUMO

A panel of influenza A viruses encoding mutant NS1 proteins was created in which a number of NS1 functions, including interactions with dsRNA, PI3K, CPSF30 and PKR, were inhibited. Surprisingly, given previous reports that NS1 activates PI3K to prevent apoptosis, the mutant viruses rUd-Y89F and rUd-P164/7A that fail to activate PI3K did not induce any more apoptosis than wild-type virus in MRC-5 and A549 cells, even though these cells are highly sensitive to inducers of apoptosis. Induction of cell death by the apoptogenic rUd-184-8(P) virus could not be prevented by serum-mediated activation of PI3K/Akt. Neither infection of MRC-5 or A549 cells with wild-type virus nor constitutive expression of NS1 prevented cell death caused by apoptosis inducers, suggesting that NS1 is not directly anti-apoptotic. Our data suggest that the loss of a functionally intact NS1 protein promotes apoptosis, but this is not due to an inability to activate PI3K.


Assuntos
Apoptose , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas não Estruturais Virais/fisiologia , Animais , Linhagem Celular , Cromonas/farmacologia , Fator de Especificidade de Clivagem e Poliadenilação/fisiologia , Ativação Enzimática , Humanos , Interferons/biossíntese , Morfolinas/farmacologia , eIF-2 Quinase/fisiologia
12.
Proc Natl Acad Sci U S A ; 106(40): 17175-80, 2009 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-19805083

RESUMO

The viruses that caused the three influenza pandemics of the twentieth century in 1918, 1957, and 1968 had distinct hemagglutinin receptor binding glycoproteins that had evolved the capacity to recognize human cell receptors. We have determined the structure of the H2 hemagglutinin from the second pandemic, the "Asian Influenza" of 1957. We compare it with the 1918 "Spanish Influenza" hemagglutinin, H1, and the 1968 "Hong Kong Influenza" hemagglutinin, H3, and show that despite its close overall structural similarity to H1, and its more distant relationship to H3, the H2 receptor binding site is closely related to that of H3 hemagglutinin. By analyzing hemagglutinins of potential H2 avian precursors of the pandemic virus, we show that the human receptor can be bound by avian hemagglutinins that lack the human-specific mutations of H2 and H3 pandemic viruses, Gln-226Leu, and Gly-228Ser. We show how Gln-226 in the avian H2 receptor binding site, together with Asn-186, form hydrogen bond networks through bound water molecules to mediate binding to human receptor. We show that the human receptor adopts a very similar conformation in both human and avian hemagglutinin-receptor complexes. We also show that Leu-226 in the receptor binding site of human virus hemagglutinins creates a hydrophobic environment near the Sia-1-Gal-2 glycosidic linkage that favors binding of the human receptor and is unfavorable for avian receptor binding. We consider the significance for the development of pandemics, of the existence of avian viruses that can bind to both avian and human receptors.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Vírus da Influenza A/metabolismo , Influenza Humana/virologia , Estrutura Secundária de Proteína , Animais , Ásia/epidemiologia , Sítios de Ligação/genética , Aves , Cristalografia por Raios X , Surtos de Doenças , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Hong Kong/epidemiologia , Humanos , Vírus da Influenza A/genética , Influenza Aviária/virologia , Influenza Humana/epidemiologia , Modelos Moleculares , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , Receptores Virais/química , Receptores Virais/metabolismo , Espanha/epidemiologia
13.
Virology ; 394(2): 321-30, 2009 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-19755201

RESUMO

A panel of eight single amino acid deletion mutants was generated within the first 24 residues of the fusion peptide domain of the of the hemagglutinin (HA) of A/Aichi/2/68 influenza A virus (H3N2 subtype). The mutant HAs were analyzed for folding, cell surface transport, cleavage activation, capacity to undergo acid-induced conformational changes, and membrane fusion activity. We found that the mutant DeltaF24, at the C-terminal end of the fusion peptide, was expressed in a non-native conformation, whereas all other deletion mutants were transported to the cell surface and could be cleaved into HA1 and HA2 to activate membrane fusion potential. Furthermore, upon acidification these cleaved HAs were able to undergo the characteristic structural rearrangements that are required for fusion. Despite this, all mutants were inhibited for fusion activity based on two separate assays. The results indicate that the mutant fusion peptide domains associate with target membranes in a non-functional fashion, and suggest that structural features along the length of the fusion peptide are likely to be relevant for optimal membrane fusion activity.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/fisiologia , Vírus da Influenza A Subtipo H3N2/genética , Vírus da Influenza A Subtipo H3N2/fisiologia , Deleção de Sequência , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/fisiologia , Internalização do Vírus , Sequência de Aminoácidos , Animais , Linhagem Celular , Cricetinae , Genes Virais , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Humanos , Concentração de Íons de Hidrogênio , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Estrutura Quaternária de Proteína , Homologia de Sequência de Aminoácidos , Proteínas Virais de Fusão/química
14.
Protein Pept Lett ; 16(7): 766-78, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19601906

RESUMO

Fusion of the influenza virus envelope with the endosomal membrane of host cells is mediated by the hemagglutinin glycoprotein (HA). The most conserved region of HA is at the N-terminus of the HA2 subunit, a relatively hydrophobic sequence of amino acids referred to as the fusion peptide. This domain is critical both for setting the trigger for fusion and for destabilizing target membranes during the fusion process. The "trigger" is set by cleavage of the HA precursor polypeptide, when the newly-generated HA2 N-terminal fusion peptide positions itself into the trimer interior and makes contacts with ionizable residues to generate a fusion competent neutral pH structure. This essentially "primes" the HA such that subsequent acidification of the endosomal environment can induce the irreversible conformational changes that result in membrane fusion. A key component of these acid-induced structural rearrangements involves the extrusion of the fusion peptide from its buried position and its relocation to interact with the target membrane. The role of the fusion peptide for both priming the neutral pH structure and interacting with cellular membranes during the fusion process is discussed.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Orthomyxoviridae/metabolismo , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Sequência de Aminoácidos , Animais , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Humanos , Concentração de Íons de Hidrogênio , Dados de Sequência Molecular , Conformação Proteica , Proteínas Virais de Fusão/genética , Internalização do Vírus
15.
J Med Chem ; 52(9): 2673-82, 2009 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-19419202

RESUMO

The tenovins and cambinol are two classes of sirtuin inhibitor that exhibit antitumor activity in preclinical models. This report describes modifications to the core structure of cambinol, in particular by incorporation of substituents at the N1-position, which lead to increased potency and modified selectivity. These improvements have been rationalized using molecular modeling techniques. The expected functional selectivity in cells was also observed for both a SIRT1 and a SIRT2 selective analog.


Assuntos
Naftalenos/química , Naftalenos/farmacologia , Pirimidinonas/química , Pirimidinonas/farmacologia , Sirtuínas/antagonistas & inibidores , Sequência de Aminoácidos , Linhagem Celular Tumoral , Humanos , Dados de Sequência Molecular , Naftalenos/síntese química , Naftalenos/metabolismo , Pirimidinonas/síntese química , Pirimidinonas/metabolismo , Sirtuínas/química , Sirtuínas/metabolismo , Especificidade por Substrato
16.
Antiviral Res ; 81(2): 132-40, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19028526

RESUMO

The antiviral drug arbidol (ARB), which is licensed in Russia for use against influenza, is known to inhibit early membrane fusion events in influenza A and B virus replication. To investigate in more detail the target and mechanism of ARB action we generated and studied the characteristics of ARB-resistant influenza virus mutants. Observations of the ARB susceptibility of reassortants between A/Singapore/1/57(H2N2) and A/chicken/Germany/27(H7N7, "Weybridge" strain) and of mutants of the latter virus identified the virus haemagglutinin (HA) as the major determinant of ARB sensitivity. ARB-resistant mutants, selected from the most sensitive reassortant, possessed single amino acid substitutions in the HA2 subunit which caused an increase in the pH of fusion and the associated conformational change in HA. ARB was shown to stabilize the HA by causing a 0.2 pH unit reduction in the pH of the transition to the low pH form, which was specifically abrogated by the resistance mutations. Some of the resistance mutations, which reduce acid stability and would disrupt ARB-HA interactions, are located in the vicinity of a potential ARB binding site identified using the docking programme Gold. Together, the results of these investigations indicate that ARB falls within a class of inhibitor which interacts with HA to stabilize it against the low pH transition to its fusogenic state and consequently inhibit HA-mediated membrane fusion during influenza virus infection.


Assuntos
Antivirais/farmacologia , Farmacorresistência Viral , Indóis/farmacologia , Vírus da Influenza A Subtipo H2N2/efeitos dos fármacos , Vírus da Influenza A Subtipo H7N7/efeitos dos fármacos , Vírus Reordenados/efeitos dos fármacos , Substituição de Aminoácidos/genética , Animais , Linhagem Celular , Cães , Hemaglutininas Virais/química , Hemaglutininas Virais/genética , Testes de Sensibilidade Microbiana , Modelos Moleculares , Mutação de Sentido Incorreto , Conformação Proteica
17.
Proc Natl Acad Sci U S A ; 105(46): 17736-41, 2008 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-19004788

RESUMO

The influenza surface glycoprotein hemagglutinin (HA) is a potential target for antiviral drugs because of its key roles in the initial stages of infection: receptor binding and the fusion of virus and cell membranes. The structure of HA in complex with a known inhibitor of membrane fusion and virus infectivity, tert-butyl hydroquinone (TBHQ), shows that the inhibitor binds in a hydrophobic pocket formed at an interface between HA monomers. Occupation of this site by TBHQ stabilizes the neutral pH structure through intersubunit and intrasubunit interactions that presumably inhibit the conformational rearrangements required for membrane fusion. The nature of the binding site suggests routes for the chemical modification of TBHQ that could lead to the development of more potent inhibitors of membrane fusion and potential anti-influenza drugs.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Hidroquinonas/química , Hidroquinonas/farmacologia , Fusão de Membrana/efeitos dos fármacos , Sítios de Ligação , Fluorometria , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Filogenia , Multimerização Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína , Subunidades Proteicas/química , Especificidade por Substrato/efeitos dos fármacos
18.
J Mol Biol ; 384(2): 436-49, 2008 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-18835278

RESUMO

The Streptococcus pneumoniae genomes encode up to three sialidases (or neuraminidases), NanA, NanB and NanC, which are believed to be involved in removing sialic acid from host cell surface glycans, thereby promoting colonization of the upper respiratory tract. Here, we present the crystal structure of NanB to 1.7 A resolution derived from a crystal grown in the presence of the buffer Ches (2-N-cyclohexylaminoethanesulfonic acid). Serendipitously, Ches was found bound to NanB at the enzyme active site, and was found to inhibit NanB with a K(i) of approximately 0.5 mM. In addition, we present the structure to 2.4 A resolution of NanB in complex with the transition-state analogue Neu5Ac2en (2-deoxy-2,3-dehydro-N-acetyl neuraminic acid), which inhibits NanB with a K(i) of approximately 0.3 mM. The sulphonic acid group of Ches and carboxylic acid group of Neu5Ac2en interact with the arginine triad of the active site. The cyclohexyl group of Ches binds in the hydrophobic pocket of NanB occupied by the acetamidomethyl group of Neu5Ac2en. The topology around the NanB active site suggests that the enzyme would have a preference for alpha2,3-linked sialoglycoconjugates, which is confirmed by a kinetic analysis of substrate binding. NMR studies also confirm this preference and show that, like the leech sialidase, NanB acts as an intramolecular trans-sialidase releasing Neu2,7-anhydro5Ac. All three pneumoccocal sialidases possess a carbohydrate-binding domain that is predicted to bind sialic acid. These studies provide support for a possible differential role for NanB compared to NanA in pneumococcal virulence.


Assuntos
Neuraminidase/química , Streptococcus pneumoniae/enzimologia , Sequência de Aminoácidos , Sítios de Ligação , Carboidratos/química , Sequência Conservada , Cristalografia por Raios X , Lectinas/metabolismo , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Dados de Sequência Molecular , Ácido N-Acetilneuramínico/análogos & derivados , Ácido N-Acetilneuramínico/metabolismo , Estrutura Terciária de Proteína , Alinhamento de Sequência , Especificidade por Substrato , Ácidos Sulfônicos/metabolismo
19.
Virology ; 378(1): 1-5, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18585749

RESUMO

Influenza A virus NS1 protein is a multifunctional virulence factor. Here, we report a crystal structure for the NS1 effector domain of avian influenza virus A/Duck/Albany/76. Comparison of this structure with that reported for a human strain shows both proteins share a common monomer conformation, albeit with subtle differences. Strikingly, our data reveal a novel helix-helix dimeric interface between monomers of the avian NS1 protein, which is also found in the human NS1 crystal lattice. We re-evaluate the current model of NS1 dimeric assembly, and provide biochemical evidence to show tryptophan-187 (a residue located at the helix-helix interface) is essential for dimerization of this effector domain.


Assuntos
Patos/virologia , Vírus da Influenza A/metabolismo , Proteínas não Estruturais Virais/química , Animais , Dicroísmo Circular , Cristalização , Cristalografia por Raios X , Dimerização , Humanos , Conformação Proteica , Estrutura Secundária de Proteína , Proteínas não Estruturais Virais/isolamento & purificação , Proteínas não Estruturais Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...