Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Commun Biol ; 3(1): 219, 2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32382088

RESUMO

Prolonged treatment of immunocompromised influenza patients with viral neuraminidase (NA) inhibitors is required, because the immune system of such patients fails to eradicate the viruses. Here, we attempted to eradicate influenza virus from the respiratory organs of nude mice, which is a model of immunocompromised hosts, by using combination therapy of the viral polymerase inhibitor favipiravir and monoclonal antibodies (mAbs) against the receptor-binding site (RBS) and stem of viral hemagglutinin (HA). Although monotherapy or combination therapy of two antivirals (two mAbs or favipiravir plus a mAb) suppressed virus replication, they failed to eradicate viruses from nude mice. In contrast, the triple combination therapy of favipiravir plus anti-Stem and anti-RBS mAbs completely stopped virus replication in nude mice, resulting in virus clearance. Triple combination approaches should be considered for the treatment of human immunocompromised patients with severe influenza.


Assuntos
Amidas/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Antivirais/administração & dosagem , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Infecções por Orthomyxoviridae/prevenção & controle , Pirazinas/administração & dosagem , Animais , Quimioterapia Combinada , Feminino , Hemaglutininas Virais/química , Hemaglutininas Virais/efeitos dos fármacos , Camundongos , Camundongos Nus
2.
Org Lett ; 20(21): 6938-6942, 2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30350678

RESUMO

Fungal glycolipids emmyguyacins A and B inhibit the pH-dependent conformational change of hemaglutinin A during replication of the Influenza virus. Herein, we report the first total synthesis and structure confirmation of emmyguyacins A and B. Our efficient route, which involves regioselective functionalization of trehalose, allows rapid access to adequate amounts of chemically pure emmyguyacin analogues including the desoxylate derivatives for SAR studies.


Assuntos
Glicolipídeos/síntese química , Hemaglutininas Virais/efeitos dos fármacos , Orthomyxoviridae/efeitos dos fármacos , Isomerismo , Estrutura Molecular , Oxalatos/química , Trealose/química
3.
Antivir Chem Chemother ; 21(4): 169-74, 2011 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-21602614

RESUMO

BACKGROUND: The recent emergence of pandemic influenza virus H1N1v stresses the need for the development of new anti-influenza drugs. METHODS: Host proteases responsible for viral haemagglutinin (HA) cleavage are attractive targets for such drugs. Aprotinin, a natural 58-amino-acid polypeptide from bovine lungs, was chosen for this purpose because it is a drug already approved for human use as an antiprotease compound to treat pancreatitis and bleeding, and because it inhibits a wide spectrum of serine proteases, some of which are involved in influenza virus activation. RESULTS: First, we show that HA of pandemic H1N1v was intensively cleaved and activated in different host systems (human tracheo-bronchial epithelium, human intestinal Caco-2 cells and chicken embryonated eggs). Second, aprotinin inhibited HA cleavage and replication of pandemic influenza virus H1N1v in all host systems, including human tracheo-bronchial epithelium. Third, aprotinin did not induce any apparent toxic side effects in these hosts. CONCLUSIONS: Aprotinin can be considered a promising drug against the novel H1N1v pandemic influenza virus.


Assuntos
Antivirais/farmacologia , Aprotinina/farmacologia , Hemaglutininas Virais/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Inibidores de Serina Proteinase/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Células CACO-2 , Bovinos , Embrião de Galinha , Hemaglutininas Virais/metabolismo , Humanos , Especificidade da Espécie
4.
Curr Pharm Des ; 17(17): 1720-39, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21619529

RESUMO

While the seasonal influenza viruses spreading around the world cause the annual epidemics, the recent outbreaks of influenza A virus subtype H5N1 and pandemic H1N1 have raised a global human health concerns. In this review, the applicability of computational techniques focused on three important targets in the viral life cycle: hemagglutinin, neuraminidase and M2 proton channel are summarized. Protein mechanism of action, substrate binding specificity and drug resistance, ligand-target interactions of substrate/inhibitor binding to these three proteins either wild-type or mutant strains are discussed and compared. Advances on the novel anti-influenza agents designed specifically to combat the avian H5N1 and pandemic H1N1 viruses are introduced. A better understanding of molecular inhibition and source of drug resistance as well as a set of newly designed compounds is greatly useful as a rotational guide for synthetic and medicinal chemists to develop a new generation of anti-influenza drugs.


Assuntos
Antivirais/farmacologia , Hemaglutininas Virais/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Neuraminidase/efeitos dos fármacos , Proteínas da Matriz Viral/efeitos dos fármacos , Biologia Computacional , Vírus da Influenza A Subtipo H1N1/enzimologia , Vírus da Influenza A Subtipo H1N1/metabolismo , Virus da Influenza A Subtipo H5N1/enzimologia , Virus da Influenza A Subtipo H5N1/metabolismo
6.
J Gen Virol ; 89(Pt 1): 60-67, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18089729

RESUMO

Influenza virus infection is one of the major causes of human morbidity and mortality. Between humans, this virus spreads mostly via aerosols excreted from the respiratory system. Current means of prevention of influenza virus infection are not entirely satisfactory because of their limited efficacy. Safe and effective preventive measures against pandemic influenza are greatly needed. We demonstrate that infection of mice induced by aerosols of influenza A virus was prevented by chlorine dioxide (ClO(2)) gas at an extremely low concentration (below the long-term permissible exposure level to humans, namely 0.1 p.p.m.). Mice in semi-closed cages were exposed to aerosols of influenza A virus (1 LD(50)) and ClO(2) gas (0.03 p.p.m.) simultaneously for 15 min. Three days after exposure, pulmonary virus titre (TCID(50)) was 10(2.6+/-1.5) in five mice treated with ClO(2), whilst it was 10(6.7+/-0.2) in five mice that had not been treated (P=0.003). Cumulative mortality after 16 days was 0/10 mice treated with ClO(2) and 7/10 mice that had not been treated (P=0.002). In in vitro experiments, ClO(2) denatured viral envelope proteins (haemagglutinin and neuraminidase) that are indispensable for infectivity of the virus, and abolished infectivity. Taken together, we conclude that ClO(2) gas is effective at preventing aerosol-induced influenza virus infection in mice by denaturing viral envelope proteins at a concentration well below the permissible exposure level to humans. ClO(2) gas could therefore be useful as a preventive means against influenza in places of human activity without necessitating evacuation.


Assuntos
Compostos Clorados/uso terapêutico , Vírus da Influenza A/efeitos dos fármacos , Influenza Humana/prevenção & controle , Óxidos/uso terapêutico , Aerossóis/farmacologia , Animais , Antibacterianos/uso terapêutico , Compostos Clorados/administração & dosagem , Hemaglutininas Virais/efeitos dos fármacos , Humanos , Vírus da Influenza A/enzimologia , Vírus da Influenza A/patogenicidade , Pulmão/efeitos dos fármacos , Pulmão/patologia , Masculino , Camundongos , Neuraminidase/efeitos dos fármacos , Óxidos/administração & dosagem , Desnaturação Proteica
7.
Antiviral Res ; 52(1): 43-53, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11530187

RESUMO

Serum amyloid P component (SAP) binds in vitro Ca(2+)-dependently to several ligands including oligosaccharides with terminal mannose and galactose. We have earlier reported that SAP binds to human influenza A virus strains, inhibiting hemagglutinin (HA) activity and virus infectivity in vitro. These studies were extended to comprise five mouse-adapted influenza A strains, two swine influenza A strains, a mink influenza A virus, a ferret influenza A reassortant virus, a influenza B virus and a parainfluenza 3 virus. The HA activity of all these viruses was inhibited by SAP. Western blotting showed that SAP bound to HA trimers, monomers and HA1 and HA2 subunits of influenza A virus. Binding studies indicated that galactose, mannose and fucose moieties contributed to the SAP reacting site(s). Intranasal administration of human SAP to mice induced no demonstrable toxic reactions, and circulating antibodies against SAP were not detected. Preincubation of virus (A/Japan/57) with SAP prevented primary infection of mice and development of antiviral antibodies. After a single intranasal administration of SAP (40 microg) 1 h before primary infection with virus (2LD(50)), nine out of 10 mice survived on day 10 and these mice approached normal body weight, whereas control mice (one out of five surviving on day 10) died. The data provide evidence of the potential of intranasally administered SAP for prophylactic treatment of influenza A virus infections in humans.


Assuntos
Antivirais/farmacologia , Hemaglutininas Virais/metabolismo , Vírus da Influenza A/efeitos dos fármacos , Infecções por Orthomyxoviridae/prevenção & controle , Componente Amiloide P Sérico/farmacologia , Animais , Antivirais/metabolismo , Compostos de Benzalcônio/farmacologia , Western Blotting , Cálcio/farmacologia , Cromatografia por Troca Iônica , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Feminino , Testes de Inibição da Hemaglutinação , Hemaglutininas Virais/química , Hemaglutininas Virais/efeitos dos fármacos , Humanos , Técnicas In Vitro , Vírus da Influenza A/química , Vírus da Influenza A/metabolismo , Vírus da Influenza B/efeitos dos fármacos , Masculino , Metilcelulose/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Vírus da Parainfluenza 3 Bovina/efeitos dos fármacos , Soroalbumina Bovina/farmacologia , Componente Amiloide P Sérico/metabolismo
8.
Bioorg Med Chem Lett ; 10(4): 341-3, 2000 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-10714495

RESUMO

A multiple sialic acid-bearing polymer 7 has been made in which a novel 4-N-substituted sialoside 5 has been coupled to polyacrylamide. The conjugate 7 has been found to inhibit the agglutination of influenza virus to red blood cells with HAI inhibition constants of around 10(-6) M, based on the sialic acid concentration.


Assuntos
Resinas Acrílicas/química , Resinas Acrílicas/farmacologia , Hemaglutininas Virais/efeitos dos fármacos , Orthomyxoviridae/química , Ácidos Siálicos/farmacologia , Resinas Acrílicas/síntese química , Animais , Galinhas/sangue , Eritrócitos/patologia , Testes de Sensibilidade Microbiana , Ácidos Siálicos/síntese química , Relação Estrutura-Atividade , Proteínas Virais/efeitos dos fármacos
9.
Arch Virol ; 144(5): 865-78, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10416371

RESUMO

Stachyflin is a novel compound having H1 and H2 subtype-specific anti-influenza A virus activity. Stachyflin has no inhibition on H3 subtype influenza A or influenza B viruses. The susceptibility of the reassortant viruses between H1 and H3 subtype influenza A viruses to Stachyflin indicated that its target was virus-encoded hemagglutinin (HA). The results of the timing of Stachyflin addition against in vitro virus infection and virus-mediated hemolysis assay suggested that the drug inhibited the HA-mediated virus-cell fusion process. More directly, Stachyflin interfered with HA conformational change induced by low pH or heat treatment. The effect of Stachyflin could not be eliminated by washing of the Stachyflin-treated virus, which caused very specific virucidal effect. This is a remarkable property among small molecules which inhibit low-pH induced HA conformational change. From these findings, we concluded that the mechanism of Stachyflin action is to inhibit HA conformational change which is necessary for virus-cell membrane fusion. Stachyflin may be used as a tool for a study of molecular mechanism of low-pH induced HA conformational change, and offers potential as a pharmaceutical agent.


Assuntos
Antivirais/farmacologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Hemaglutininas Virais/química , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza B/efeitos dos fármacos , Sesquiterpenos/farmacologia , Animais , Linhagem Celular , Galinhas , Cricetinae , Cães , Células Gigantes/efeitos dos fármacos , Glicoproteínas de Hemaglutininação de Vírus da Influenza/efeitos dos fármacos , Hemaglutininas Virais/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Humanos , Vírus da Influenza A/classificação , Vírus da Influenza A/fisiologia , Vírus da Influenza B/classificação , Vírus da Influenza B/fisiologia , Fusão de Membrana/efeitos dos fármacos , Conformação Proteica/efeitos dos fármacos
10.
J Virol ; 73(1): 140-51, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9847316

RESUMO

Several compounds that specifically inhibited replication of the H1 and H2 subtypes of influenza virus type A were identified by screening a chemical library for antiviral activity. In single-cycle infections, the compounds inhibited virus-specific protein synthesis when added before or immediately after infection but were ineffective when added 30 min later, suggesting that an uncoating step was blocked. Sequencing of hemagglutinin (HA) genes of several independent mutant viruses resistant to the compounds revealed single amino acid changes that clustered in the stem region of the HA trimer in and near the HA2 fusion peptide. One of the compounds, an N-substituted piperidine, could be docked in a pocket in this region by computer-assisted molecular modeling. This compound blocked the fusogenic activity of HA, as evidenced by its inhibition of low-pH-induced cell-cell fusion in infected cell monolayers. An analog which was more effective than the parent compound in inhibiting virus replication was synthesized. It was also more effective in blocking other manifestations of the low-pH-induced conformational change in HA, including virus inactivation, virus-induced hemolysis of erythrocytes, and susceptibility of the HA to proteolytic degradation. Both compounds inhibited viral protein synthesis and replication more effectively in cells infected with a virus mutated in its M2 protein than with wild-type virus. The possible functional relationship between M2 and HA suggested by these results is discussed.


Assuntos
Antivirais/farmacologia , Fusão Celular/efeitos dos fármacos , Hemaglutininas Virais/efeitos dos fármacos , Vírus da Influenza A/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Amantadina/farmacologia , Animais , Linhagem Celular , Clonagem Molecular , Simulação por Computador , Concentração de Íons de Hidrogênio , Vírus da Influenza A/fisiologia , Modelos Moleculares , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Virais/biossíntese
11.
Scand J Immunol ; 46(4): 331-7, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9350282

RESUMO

Serum amyloid P component (SAP) is a member of the phylogenetically conserved and structurally related group of proteins called pentraxins. SAP exhibits multispecific calcium-dependent binding to oligosaccharides with terminal N-acetyl-galactosamine, mannose and glucuronic acid. The authors report that SAP can bind to influenza A virus and inhibit agglutination of erythrocytes mediated by the virus subtypes H1N1, H2N2 and H3N2. SAP also inhibits the production of haemagglutinin (HA) an the cytopathogenic effect of influenza A virus in MDCK cells. The binding of SAP to the virus requires physiological calcium concentrations and is blocked by specific SAP antibodies. Denaturated and renaturated SAP retained inhibition of HA. Electron microscopy shows Ca(2+)-dependent binding of SAP to spikes on the viral envelope and immunoblotting indicates that SAP binds to a 50-55 kDa peptide corresponding to the mass of the HA1 peptide. Of several monosaccharides tested only D-mannose interfered with SAP's inhibition of both HA and infectivity. The glycosaminoglycans heparan sulfate and heparin, which bind SAP, reduced SAPs binding to the virus. The results indicate that the inhibition by SAP is due to steric effects when SAP binds to terminal mannose on oligosaccharides localized close to the sialic acid-binding site of the HA trimer.


Assuntos
Antivirais/farmacologia , Hemaglutininas Virais/metabolismo , Vírus da Influenza A/metabolismo , Infecções por Orthomyxoviridae/prevenção & controle , Componente Amiloide P Sérico/metabolismo , Componente Amiloide P Sérico/fisiologia , Acetilgalactosamina/farmacologia , Animais , Antivirais/metabolismo , Western Blotting , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Cães , Glicosaminoglicanos/farmacologia , Testes de Inibição da Hemaglutinação , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Hemaglutininas Virais/efeitos dos fármacos , Humanos , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/ultraestrutura , Rim , Monossacarídeos/farmacologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Ligação Proteica , Desnaturação Proteica , Componente Amiloide P Sérico/ultraestrutura
12.
Res Vet Sci ; 62(3): 261-4, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9300545

RESUMO

Heparin inhibited haemagglutination by porcine reproductive and respiratory syndrome virus (PRRSV) and by Aujesky's disease virus, but failed to inhibit haemagglutination by parainfluenza virus type 3. The minimal inhibitory concentration of heparin required to inhibit 8 HA U of PRRSV haemagglutinin ranged from 0.1 to 1 U ml-1. Mouse erythrocytes failed to combine with the haemagglutination inhibitory factor of heparin. However, mouse erythrocytes treated with heparinase had greatly reduced agglutinability by PRRSV. The formation of a haemagglutinin-heparin complex could be observed by sedimenting heparin with the haemagglutinin. All these findings suggest that a heparin-like molecule on the surface of mouse erythrocytes serves as the virus-cell receptor.


Assuntos
Hemaglutinação por Vírus/efeitos dos fármacos , Hemaglutininas Virais/efeitos dos fármacos , Heparina/farmacologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/efeitos dos fármacos , Animais , Bovinos , Linhagem Celular , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Eritrócitos/citologia , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Hemaglutininas Virais/fisiologia , Heparina/metabolismo , Heparina Liase/farmacologia , Herpesvirus Suídeo 1/efeitos dos fármacos , Herpesvirus Suídeo 1/fisiologia , Rim/citologia , Rim/efeitos dos fármacos , Rim/metabolismo , Camundongos , Vírus da Síndrome Respiratória e Reprodutiva Suína/fisiologia , Respirovirus/efeitos dos fármacos , Respirovirus/fisiologia , Suínos
13.
J Biol Chem ; 271(23): 13724-30, 1996 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-8662732

RESUMO

Surfactant protein D (SP-D) molecules are preferentially assembled as dodecamers consisting of trimeric subunits associated at their amino termini. The NH2-terminal sequence of each monomer contains two conserved cysteine residues, which participate in interchain disulfide bonds. In order to study the roles of these residues in SP-D assembly and function, we employed site-directed mutagenesis to substitute serine for cysteine 15 and 20 in recombinant rat SP-D (RrSP-D), and have expressed the mutant (RrSP-Dser15/20) in Chinese hamster ovary (CHO-K1) cells. The mutant, which was efficiently secreted, bound to maltosyl-agarose, but unlike RrSP-D, was assembled exclusively as trimers. The constituent monomers showed a decreased mobility on SDS-polyacrylamide gel electrophoresis resulting from an increase in the size and sialylation of the N-linked oligosaccharide at Asn-70. Although RrSP-Dser15/20 contained a pepsin-resistant triple helical domain, it showed a decreased Tm, and acquired susceptibility to proteolytic degradation. Like RrSP-D, RrSP-Dser15/20 bound to the hemagglutinin of influenza A. However, it showed no viral aggregation and did not enhance the binding of influenza A to neutrophils (PMN), augment PMN respiratory burst, or protect PMNs from deactivation. These studies indicate that amino-terminal disulfides are required to stabilize dodecamers, and support our hypothesis that the oligomerization of trimeric subunits contributes to the anti-microbial properties of SP-D.


Assuntos
Antivirais/farmacologia , Glicoproteínas/genética , Glicoproteínas/farmacologia , Surfactantes Pulmonares/genética , Surfactantes Pulmonares/farmacologia , Animais , Antivirais/química , Sequência de Bases , Células CHO , Cricetinae , Cisteína/química , Cisteína/genética , Primers do DNA/genética , Glicoproteínas/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Hemaglutininas Virais/efeitos dos fármacos , Hemaglutininas Virais/metabolismo , Humanos , Técnicas In Vitro , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Conformação Proteica , Processamento de Proteína Pós-Traducional , Proteína D Associada a Surfactante Pulmonar , Surfactantes Pulmonares/química , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia
14.
Microbiol Immunol ; 40(5): 365-71, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8805101

RESUMO

The hemagglutinating (HA) activity of extracellular and intracellular forms of Japanese encephalitis (JE) virus was comparatively titrated by exposure to acidic pH below 7.0. A pH-dependent irreversible loss in titer was observed with the virus grown in both C6/36 and BHK 21 (BHK) cells maintained in the pH range of 5.8 to 7.0 for 10 min at 37 C. The HA activity of intracellular virus was relatively more stable than that of extracellular virus in the pH range of 5.8 to 6.4. Virion structural components, envelope glycoprotein (E), capsid (C), and membrane (M) proteins in extracellular virus and E, C, and the precursor form of M (prM) proteins in intracellular virus were detected by SDS-polyacrylamide gel electrophoresis (SDS-PAGE) and immunoblotting. A panel of monoclonal antibody (mAb) directed for nine antigenic epitopes on the JE virus E protein molecule was used for the analysis of antigenic reactivity of E protein after treatment at pH 6.0. The reaction between the extracellular virus and three HA-inhibiting (HI) mAbs was significantly reduced after acid treatment; however, the antigenic reactivity of intracellular virus was much more stable with a 100- to 1,000-fold difference. Infectivity titers of extracellular and intracellular viruses in Vero cells were reduced by 1/24,100 and 1/21,666 after acidic treatment at pH 6.0. In contrast, the infectivity of intracellular viruses was more stable, with residual infectivity of 1/182 and 1/340 for BHK and C6/36 cell-grown virus, respectively. Acidic treatment of JE virus not only resulted in the irreversible loss of its HA activity but also affected the antigenic reactivity of HI epitopes on its E protein molecule.


Assuntos
Ácidos/toxicidade , Vírus da Encefalite Japonesa (Espécie)/classificação , Vírus da Encefalite Japonesa (Espécie)/efeitos dos fármacos , Hemaglutininas Virais/efeitos dos fármacos , Hemaglutininas Virais/metabolismo , Animais , Linhagem Celular , Cricetinae , Culicidae , Vírus da Encefalite Japonesa (Espécie)/metabolismo , Vírus da Encefalite Japonesa (Espécie)/patogenicidade , Rim/citologia
15.
Biochem Biophys Res Commun ; 214(2): 318-23, 1995 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-7677737

RESUMO

Fractions obtained from pine cone extract (PCE) of Pinus parviflora Sieb. et Zucc. have been shown to suppress the growth of influenza virus. The inhibitory effects of one of the fractions, Fraction VII, on the formation of RNA-viral protein complex and the viral RNA synthesis were investigated. The formation of M1-RNA or NP-RNA complex was inhibited when M1 or NP was preincubated with the PCE fraction. The in vitro viral RNA synthesis was inhibited by the PCE fraction, while this inhibitory effect was titrated out by the increasing concentration of M1 protein. These results suggest that the major target of the PCE fraction was M1 protein.


Assuntos
Antineoplásicos/farmacologia , Gammainfluenzavirus/fisiologia , Hemaglutininas Virais/metabolismo , Extratos Vegetais/farmacologia , RNA Viral/metabolismo , Árvores , Proteínas Virais de Fusão , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos , Antineoplásicos/metabolismo , Sequência de Bases , Capsídeo/biossíntese , Capsídeo/isolamento & purificação , Capsídeo/metabolismo , Clonagem Molecular , Primers do DNA , Glicoproteínas/metabolismo , Hemaglutininas Virais/biossíntese , Hemaglutininas Virais/efeitos dos fármacos , Histidina , Gammainfluenzavirus/efeitos dos fármacos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , RNA Viral/efeitos dos fármacos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Sitios de Sequências Rotuladas , Transcrição Gênica/efeitos dos fármacos , Proteínas do Core Viral/biossíntese , Proteínas do Core Viral/isolamento & purificação , Proteínas do Core Viral/metabolismo , Proteínas Virais/biossíntese , Proteínas Virais/efeitos dos fármacos
16.
Virology ; 210(1): 20-8, 1995 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-7793071

RESUMO

Influenza virus hemagglutinin (HA) subtype H7 expressed from a baculovirus vector in insect cells requires cysteine residues for palmitoylation. Mutant HA devoid of fatty acids shows hemagglutinating and hemolytic activities almost identical to those of the acylated wild-type HA (wt). Using a membrane mixing assay (R18), neither the kinetics nor the pH dependence of fusion induced by wt or mutant HA was significantly different from virus-induced fusion. HA-induced fusion of insect cells with human erythrocyte ghosts could also be demonstrated by a cytoplasmic content mixing assay. Both species of recombinant HA induced the flow of lucifer yellow from preloaded ghosts into the cytoplasm of HA-bearing cells. This indicates that membrane fusion mediated by wild-type and fatty-acid-free HA includes both leaflets of the lipid bilayers. Hydroxylamine treatment of wt HA (H7) and fatty-acid-free mutant HA present in lysates of insect cells led to the complete inhibition of hemolytic activity. Deacylation of spike proteins by NH2OH treatment of virus particles resulted in a block of hemolytic activity in influenza virus subtypes H7 and H10 as well as of that in the togaviruses Semliki Forest and Sindbis virus. However, the same treatment did not affect subtypes H2 and H3 or two vesicular stomatitis virus serotypes. With such a differential effect whether or not fatty acids are present in the spike proteins of the different virus particles, hydroxylamine must have other effects than just deacylation, and therefore seems unsuitable for the study of the biological functions of acylproteins.


Assuntos
Fusão Celular , Membrana Eritrocítica/fisiologia , Hemaglutininas Virais/fisiologia , Hidroxilaminas/farmacologia , Vírus da Influenza A/imunologia , Acilação , Sequência de Aminoácidos , Animais , Linhagem Celular , Membrana Eritrocítica/imunologia , Hemaglutininas Virais/biossíntese , Hemaglutininas Virais/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio , Hidroxilamina , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Spodoptera , Transfecção
17.
J Virol ; 68(2): 911-9, 1994 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7507186

RESUMO

The influenza A/fowl plague virus/Rostock/34 hemagglutinin (HA), which is cleaved intracellularly and has a high pH threshold (pH 5.9) for undergoing its conformational change to the low-pH form, was expressed from cDNA in CV-1 and HeLa T4 cells in the absence of other influenza virus proteins. It was found, by biochemical assays, that the majority of the HA molecules were in a form indistinguishable from the low-pH form of HA. The acidotropic agent, ammonium chloride, stabilized the accumulation of HA in its native form. Coexpression of HA and the homotypic influenza virus M2 protein, which has ion channel activity, stabilized the accumulation of HA in its pH neutral (native) form, and the M2 protein ion channel blocker, amantadine, prevented the rescue of HA in its native form. These data provide direct evidence that the influenza virus M2 protein ion channel activity can affect the status of the conformational form of cleaved HA during intracellular transport.


Assuntos
Hemaglutininas Virais/metabolismo , Vírus da Influenza A/metabolismo , Canais Iônicos , Proteínas da Matriz Viral/metabolismo , Amantadina/farmacologia , Cloreto de Amônio/farmacologia , Animais , Transporte Biológico , Células Cultivadas , Chlorocebus aethiops , Endopeptidases/farmacologia , Células HeLa , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Hemaglutininas Virais/efeitos dos fármacos , Hemaglutininas Virais/genética , Hexosaminidases/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Oxirredução , Conformação Proteica , Proteínas Recombinantes/metabolismo , Proteínas da Matriz Viral/genética
18.
Arch Virol ; 135(1-2): 209-16, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-7515225

RESUMO

Chicken embryo proteinases, one of which is a blood clotting factor Xa-like proteinase, are known to effectively cleave the haemagglutinin (HA) of Influenza B viruses to permit their replication in chicken embryonated eggs. Here we show that injection of the serine proteinase inhibitor, aprotinin, into the allantoic cavity of eggs infected with Influenza B/Hong Kong/73 and B/Lee/40 viruses suppresses the viral HA cleavage and reduces the virus proteolytic activation and replication. Effective inhibition dose was determined as approximately 10.0 micrograms of aprotinin per embryo that corresponds to 0.1 microM concentration. However, heparin, which is known to be a direct inhibitor of the Factor Xa, was not able to suppress Influenza B virus hemagglutinin cleavage and replication in chicken embryo system. These data shed light on the pattern of proteinases involved in the Influenza B virus proteolytic activation and indicate that aprotinin possesses antiviral potential against Influenza B viruses.


Assuntos
Antivirais/farmacologia , Aprotinina/farmacologia , Vírus da Influenza B/efeitos dos fármacos , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos , Animais , Embrião de Galinha , Eletroforese em Gel de Poliacrilamida , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Hemaglutininas Virais/efeitos dos fármacos , Hemaglutininas Virais/metabolismo , Vírus da Influenza B/crescimento & desenvolvimento , Vírus da Influenza B/fisiologia , Especificidade da Espécie , Proteínas do Envelope Viral/metabolismo , Ensaio de Placa Viral , Proteínas Virais/isolamento & purificação , Ativação Viral/efeitos dos fármacos
19.
Microbiol Immunol ; 37(12): 979-81, 1993.
Artigo em Inglês | MEDLINE | ID: mdl-8133804

RESUMO

Heparin inhibited the hemagglutinin activity of herpes simplex virus (HSV) type 1. The minimal inhibitory concentration of heparin required to inhibit 8 hemagglutination (HA) U of HSV ranged from 0.005 to 0.01 U/ml. Mouse erythrocytes failed to combine with the HA inhibitory factor of heparin. On the other hand, mouse erythrocytes treated with heparinase had greatly reduced agglutinability by HSV. Virus-heparin complex formation was observed by sedimenting heparin with the virus particles.


Assuntos
Hemaglutininas Virais/efeitos dos fármacos , Heparina/farmacologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/imunologia , Animais , Testes de Inibição da Hemaglutinação , Testes de Hemaglutinação , Camundongos
20.
Virology ; 188(1): 14-24, 1992 May.
Artigo em Inglês | MEDLINE | ID: mdl-1566569

RESUMO

Amantadine treatment of cells infected with H7 strains of influenza A viruses causes an M2 protein-mediated conversion of hemagglutinin (HA) from its native to its low pH conformation. Immunofluorescence and electron microscopic observations showed that the structural alteration and hence drug action occur shortly after HA exits from the Golgi complex during its passage through the strans Golgi region. Using the DAMP/anti-DNP pH probe it is evident that virus infection causes increased acidity of the trans Golgi region and that vesicles containing low pH HA in amantadine-treated virus-infected cells are particularly acidic. These results indicate therefore that the alteration in HA is the direct consequence of exposure to an adverse low pH and provide further support for the conclusion that the M2 protein, the target of amantadine action, is involved in regulating vesicular pH, a function important for the correct maturation of the HA glycoprotein.


Assuntos
Amantadina/farmacologia , Complexo de Golgi/metabolismo , Hemaglutininas Virais/química , Vírus da Influenza A/efeitos dos fármacos , Animais , Compartimento Celular , Células Cultivadas , Imunofluorescência , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Hemaglutininas Virais/efeitos dos fármacos , Hemaglutininas Virais/metabolismo , Concentração de Íons de Hidrogênio , Vírus da Influenza A/ultraestrutura , Microscopia Imunoeletrônica , Monensin/farmacologia , Conformação Proteica/efeitos dos fármacos , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...