Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Cell Physiol ; 319(3): C500-C509, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32687406

RESUMO

Severe acute respiratory syndrome coronavirus (SARS-CoV), an enveloped virus with a positive-sense single-stranded RNA genome, facilitates the host cell entry through intricate interactions with proteins and lipids of the cell membrane. The detailed molecular mechanism involves binding to the host cell receptor and fusion at the plasma membrane or after being trafficked to late endosomes under favorable environmental conditions. A crucial event in the process is the proteolytic cleavage of the viral spike protein by the host's endogenous proteases that releases the fusion peptide enabling fusion with the host cellular membrane system. The present review details the mechanism of viral fusion with the host and highlights the therapeutic options that prevent SARS-CoV-2 entry in humans.


Assuntos
Betacoronavirus/metabolismo , Membrana Celular/metabolismo , Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/prevenção & controle , Pandemias/prevenção & controle , Pneumonia Viral/metabolismo , Pneumonia Viral/prevenção & controle , Proteínas Virais de Fusão/metabolismo , Sequência de Aminoácidos , Enzima de Conversão de Angiotensina 2 , Animais , Betacoronavirus/efeitos dos fármacos , COVID-19 , Membrana Celular/efeitos dos fármacos , Membrana Celular/virologia , Humanos , Peptidil Dipeptidase A/metabolismo , Inibidores de Proteases/farmacologia , Inibidores de Proteases/uso terapêutico , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/antagonistas & inibidores , Glicoproteína da Espícula de Coronavírus/metabolismo , Proteínas Virais de Fusão/efeitos dos fármacos
2.
Viruses ; 11(7)2019 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-31288455

RESUMO

BACKGROUND: To date, there is no licensed vaccine available to prevent respiratory syncytial virus (RSV) infection. The valuable pre-fusion conformation of the fusion protein (pre-F) is prone to lose high neutralizing antigenic sites. The goals of this study were to stabilize pre-F protein by fixatives and try to find the possibility of developing an inactivated RSV vaccine. METHODS: The screen of the optimal fixative condition was performed with flow cytometry. BALB/c mice were immunized intramuscularly with different immunogens. The serum neutralizing antibody titers of immunized mice were determined by neutralization assay. The protection and safety of these immunogens were assessed. RESULTS: Fixation in an optimal concentration of formaldehyde (0.0244%-0.0977%) or paraformaldehyde (0.0625%-1%) was able to stabilize pre-F. Additionally, BALB/c mice inoculated with optimally stabilized pre-F protein (opti-fixed) induced a higher anti-RSV neutralization (9.7 log2, mean value of dilution rate) than those inoculated with unstable (unfixed, 8.91 log2, p < 0.01) or excessively fixed (exce-fixed, 7.28 log2, p < 0.01) pre-F protein. Furthermore, the opti-fixed immunogen did not induce enhanced RSV disease. CONCLUSIONS: Only the proper concentration of fixatives could stabilize pre-F and the optimal formaldehyde condition provides a potential reference for development of an inactivated RSV vaccine.


Assuntos
Formaldeído/farmacologia , Vírus Sinciciais Respiratórios/metabolismo , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/efeitos dos fármacos , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Linhagem Celular , Modelos Animais de Doenças , Epitopos , Imunização , Imunoglobulina G , Camundongos , Camundongos Endogâmicos BALB C , Conformação Proteica , Infecções por Vírus Respiratório Sincicial/imunologia , Vacinação , Vacinas de Produtos Inativados
3.
J Virol ; 93(17)2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31217248

RESUMO

Enveloped viruses utilize surface glycoproteins to bind and fuse with a target cell membrane. The zoonotic Hendra virus (HeV), a member of the family Paramyxoviridae, utilizes the attachment protein (G) and the fusion protein (F) to perform these critical functions. Upon triggering, the trimeric F protein undergoes a large, irreversible conformation change to drive membrane fusion. Previously, we have shown that the transmembrane (TM) domain of the F protein, separate from the rest of the protein, is present in a monomer-trimer equilibrium. This TM-TM association contributes to the stability of the prefusion form of the protein, supporting a role for TM-TM interactions in the control of F protein conformational changes. To determine the impact of disrupting TM-TM interactions, constructs expressing the HeV F TM with limited flanking sequences were synthesized. Coexpression of these constructs with HeV F resulted in dramatic reductions in the stability of F protein expression and fusion activity. In contrast, no effects were observed when the HeV F TM constructs were coexpressed with the nonhomologous parainfluenza virus 5 (PIV5) fusion protein, indicating a requirement for specific interactions. To further examine this, a TM peptide homologous to the PIV5 F TM domain was synthesized. Addition of the peptide prior to infection inhibited infection with PIV5 but did not significantly affect infection with human metapneumovirus, a related virus. These results indicate that targeted disruption of TM-TM interactions significantly impact viral fusion protein stability and function, presenting these interactions as a novel target for antiviral development.IMPORTANCE Enveloped viruses require virus-cell membrane fusion to release the viral genome and replicate. The viral fusion protein triggers from the pre- to the postfusion conformation, an essentially irreversible change, to drive membrane fusion. We found that small proteins containing the TM and a limited flanking region homologous to the fusion protein of the zoonotic Hendra virus reduced protein expression and fusion activity. The introduction of exogenous TM peptides may displace a TM domain, disrupting native TM-TM interactions and globally destabilizing the fusion protein. Supporting this hypothesis, we showed that a sequence-specific transmembrane peptide dramatically reduced viral infection in another enveloped virus model, suggesting a broader inhibitory mechanism. Viral fusion protein TM-TM interactions are important for protein function, and disruption of these interactions dramatically reduces protein stability.


Assuntos
Paramyxovirinae/metabolismo , Peptídeos/farmacologia , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética , Animais , Sítios de Ligação/efeitos dos fármacos , Chlorocebus aethiops , Vírus Hendra/química , Vírus Hendra/genética , Vírus Hendra/metabolismo , Interações Hidrofóbicas e Hidrofílicas/efeitos dos fármacos , Vírus da Parainfluenza 5/química , Vírus da Parainfluenza 5/genética , Vírus da Parainfluenza 5/metabolismo , Paramyxovirinae/química , Paramyxovirinae/genética , Conformação Proteica/efeitos dos fármacos , Domínios Proteicos/efeitos dos fármacos , Estabilidade Proteica , Células Vero , Proteínas Virais de Fusão/efeitos dos fármacos
4.
Viruses ; 11(5)2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-31096572

RESUMO

Identifying drug resistance mutations is important for the clinical use of antivirals and can help define both a drug's mechanism of action and the mechanistic basis of resistance. Resistance mutations are often identified one-at-a-time by studying viral evolution within treated patients or during viral growth in the presence of a drug in cell culture. Such approaches have previously mapped resistance to enfuvirtide, the only clinically approved HIV-1 fusion inhibitor, to enfuvirtide's binding site in the N-terminal heptad repeat (NHR) of the Envelope (Env) transmembrane domain as well as a limited number of allosteric sites. Here, we sought to better delineate the genotypic determinants of resistance throughout Env. We used deep mutational scanning to quantify the effect of all single-amino-acid mutations to the subtype A BG505 Env on resistance to enfuvirtide. We identified both previously characterized and numerous novel resistance mutations in the NHR. Additional resistance mutations clustered in other regions of Env conformational intermediates, suggesting they may act during different fusion steps by altering fusion kinetics and/or exposure of the enfuvirtide binding site. This complete map of resistance sheds light on the diverse mechanisms of enfuvirtide resistance and highlights the utility of using deep mutational scanning to comprehensively map potential drug resistance mutations.


Assuntos
Antirretrovirais/farmacologia , Farmacorresistência Viral/efeitos dos fármacos , Farmacorresistência Viral/genética , Inibidores da Fusão de HIV/farmacologia , HIV-1/efeitos dos fármacos , HIV-1/genética , Enfuvirtida/farmacologia , Genótipo , Proteína gp41 do Envelope de HIV , Infecções por HIV/tratamento farmacológico , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Modelos Moleculares , Mutação , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/efeitos dos fármacos
5.
Antiviral Res ; 167: 68-77, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30953674

RESUMO

Lassa virus (LASV) causes Lassa hemorrhagic fever in humans and poses a significant threat to public health in West Africa. Current therapeutic treatments for Lassa fever are limited, making the development of novel countermeasures an urgent priority. In this study, we identified losmapimod, a p38 mitogen-activated protein kinase (MAPK) inhibitor, from 102 screened compounds as an inhibitor of LASV infection. Losmapimod exerted its inhibitory effect against LASV after p38 MAPK down-regulation, and, interestingly, had no effect on other arenaviruses capable of causing viral hemorrhagic fever. Mechanistic studies showed that losmapimod inhibited LASV entry by affecting the stable signal peptide (SSP)-GP2 subunit interface of the LASV glycoprotein, thereby blocking pH-dependent viral fusion. As an aryl heteroaryl bis-carboxyamide derivative, losmapimod represents a novel chemical scaffold with anti-LASV activity, and it provides a new lead structure for the future development of LASV fusion inhibitors.


Assuntos
Antivirais/farmacologia , Ciclopropanos/farmacologia , Vírus Lassa/efeitos dos fármacos , Piridinas/farmacologia , Internalização do Vírus/efeitos dos fármacos , Animais , Infecções por Arenaviridae/tratamento farmacológico , Arenavirus/efeitos dos fármacos , Linhagem Celular , Chlorocebus aethiops , Reposicionamento de Medicamentos , Inibidores Enzimáticos/farmacologia , Humanos , Febre Lassa/tratamento farmacológico , Febre Lassa/virologia , Células Vero , Proteínas Virais de Fusão/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
Drug Discov Ther ; 11(5): 246-252, 2017 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-29070744

RESUMO

The effect of glycyrrhizin on the replication of human parainfluenza virus type 2 (hPIV-2) was examined. Cell fusion induced by hPIV-2 was inhibited by glycyrrhizin, and glycyrrhizin reduced the number of viruses released from the cells. Glycyrrhizin did not change cell morphology at 1 day of culture, but caused some damage at 4 days, as determined by the effect on actin microfilaments. However, it affected the cell viability at 1 day: about 20% of the cells were not alive by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay at 1 day of culture. Real-time polymerase chain reaction (PCR) and PCR showed that virus genome synthesis was largely inhibited. mRNA synthesis was also inhibited by glycyrrhizin. Viral protein synthesis was largely inhibited as observed by an indirect immunofluorescence study. Multinucleated giant cell formation was studied using a recombinant green fluorescence protein (GFP)-expressing hPIV-2 without matrix protein (rhPIV-2ΔMGFP). A few single cells with fluorescence were observed, but the formation of giant cells was completely blocked. Taken together, it was shown that viral genome, mRNA and protein syntheses, including F and HN proteins, were inhibited by glycyrrhizin, and consequently multinucleated giant cell formation was not observed and the infectious virus was not detected in the culture medium.


Assuntos
Ácido Glicirrízico/farmacologia , Vírus da Parainfluenza 2 Humana/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , RNA Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Citoesqueleto de Actina/efeitos dos fármacos , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Genoma Viral/efeitos dos fármacos , Células Gigantes/efeitos dos fármacos , Proteína HN/biossíntese , Proteína HN/efeitos dos fármacos , Macaca mulatta , Vírus da Parainfluenza 2 Humana/genética , Vírus da Parainfluenza 2 Humana/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Virais de Fusão/biossíntese , Proteínas Virais de Fusão/efeitos dos fármacos , Proteínas Virais/biossíntese , Proteínas Virais/efeitos dos fármacos , Replicação Viral/genética
7.
Adv Exp Med Biol ; 966: 37-54, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27966108

RESUMO

The influenza virus is a major health concern associated with an estimated 5000 to 30,000 deaths every year (Reed et al. 2015) and a significant economic impact with the development of treatments, vaccinations and research (Molinari et al. 2007). The entirety of the influenza genome is comprised of only eleven coding genes. An enormous degree of variation in non-conserved regions leads to significant challenges in the development of inclusive inhibitors for treatment. The fusion peptide domain of the influenza A hemagglutinin (HA) is a promising candidate for treatment since it is one of the most highly conserved sequences in the influenza genome (Heiny et al. 2007), and it is vital to the viral life cycle. Hemagglutinin is a class I viral fusion protein that catalyzes the membrane fusion process during cellular entry and infection. Impediment of the hemagglutinin's function, either through incomplete post-translational processing (Klenk et al. 1975; Lazarowitz and Choppin 1975) or through mutations (Cross et al. 2001), leads to non-infective virus particles. This review will investigate current research on the role of hemagglutinin in the virus life cycle, its structural biology and mechanism as well as the central role of the hemagglutinin fusion peptide (HAfp) to influenza membrane fusion and infection.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Vírus da Influenza A/metabolismo , Influenza Humana/virologia , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus , Animais , Desenho de Fármacos , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/efeitos dos fármacos , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/patogenicidade , Influenza Humana/tratamento farmacológico , Modelos Moleculares , Domínios Proteicos , Relação Estrutura-Atividade , Inibidores de Proteínas Virais de Fusão/química , Inibidores de Proteínas Virais de Fusão/uso terapêutico , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/efeitos dos fármacos , Virulência , Internalização do Vírus/efeitos dos fármacos
8.
Hepatology ; 63(1): 49-62, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26248546

RESUMO

UNLABELLED: To explore mechanisms of hepatitis C viral (HCV) replication we screened a compound library including licensed drugs. Flunarizine, a diphenylmethylpiperazine used to treat migraine, inhibited HCV cell entry in vitro and in vivo in a genotype-dependent fashion. Analysis of mosaic viruses between susceptible and resistant strains revealed that E1 and E2 glycoproteins confer susceptibility to flunarizine. Time of addition experiments and single particle tracking of HCV demonstrated that flunarizine specifically prevents membrane fusion. Related phenothiazines and pimozide also inhibited HCV infection and preferentially targeted HCV genotype 2 viruses. However, phenothiazines and pimozide exhibited improved genotype coverage including the difficult to treat genotype 3. Flunarizine-resistant HCV carried mutations within the alleged fusion peptide and displayed cross-resistance to these compounds, indicating that these drugs have a common mode of action. CONCLUSION: These observations reveal novel details about HCV membrane fusion; moreover, flunarizine and related compounds represent first-in-class HCV fusion inhibitors that merit consideration for repurposing as a cost-effective component of HCV combination therapies.


Assuntos
Flunarizina/farmacologia , Hepacivirus/efeitos dos fármacos , Proteínas Virais de Fusão/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Células Cultivadas , Genótipo , Hepacivirus/genética , Humanos , Proteínas Virais de Fusão/genética
10.
Nat Commun ; 6: 7688, 2015 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-26158910

RESUMO

The high pathogenicity of the Ebola virus reflects multiple concurrent processes on infection. Among other important determinants, Ebola fusogenic glycoprotein (GP) has been associated with the detachment of infected cells and eventually leads to vascular leakage and haemorrhagic fever. Here we report that the membrane-anchored GP is sufficient to induce the detachment of adherent cells. The results show that the detachment induced through either full-length GP1,2 or the subunit GP2 depends on cholesterol and the structure of the transmembrane domain. These data reveal a novel molecular mechanism in which GP regulates Ebola virus assembly and suggest that cholesterol-reducing agents could be useful as therapeutics to counteract GP-mediated cell detachment.


Assuntos
Anticolesterolemiantes/farmacologia , Adesão Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Colesterol/metabolismo , Citocalasina B/farmacologia , Ebolavirus/efeitos dos fármacos , Sinvastatina/farmacologia , Proteínas Virais de Fusão/efeitos dos fármacos , beta-Ciclodextrinas/farmacologia , Animais , Células COS , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Chlorocebus aethiops , Ebolavirus/metabolismo , Ebolavirus/patogenicidade , Citometria de Fluxo , Células HEK293 , Células HeLa , Células Endoteliais da Veia Umbilical Humana , Humanos , Microscopia Confocal , Estrutura Terciária de Proteína , Proteínas Virais de Fusão/metabolismo , Fatores de Virulência/metabolismo
11.
J Pept Sci ; 21(5): 379-86, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25331523

RESUMO

While it is now possible to identify and genetically fingerprint the causative agents of emerging viral diseases, often with extraordinary speed, suitable therapies cannot be developed with equivalent speed, because drug discovery requires information that goes beyond knowledge of the viral genome. Peptides, however, may represent a special opportunity. For all enveloped viruses, fusion between the viral and the target cell membrane is an obligatory step of the life cycle. Class I fusion proteins harbor regions with a repeating pattern of amino acids, the heptad repeats (HRs), that play a key role in fusion, and HR-derived peptides such as enfuvirtide, in clinical use for HIV, can block the process. Because of their characteristic sequence pattern, HRs are easily identified in the genome by means of computer programs, providing the sequence of candidate peptide inhibitors directly from genomic information. Moreover, a simple chemical modification, the attachment of a cholesterol group, can dramatically increase the antiviral potency of HR-derived inhibitors and simultaneously improve their pharmacokinetics. Further enhancement can be provided by dimerization of the cholesterol-conjugated peptide. The examples reported so far include inhibitors of retroviruses, paramyxoviruses, orthomyxoviruses, henipaviruses, coronaviruses, and filoviruses. For some of these viruses, in vivo efficacy has been demonstrated in suitable animal models. The combination of bioinformatic lead identification and potency/pharmacokinetics improvement provided by cholesterol conjugation may form the basis for a rapid response strategy, where development of an emergency cholesterol-conjugated therapeutic would immediately follow the availability of the genetic information of a new enveloped virus.


Assuntos
Colesterol/química , Peptídeos/química , Peptídeos/farmacocinética , Viroses/tratamento farmacológico , Animais , Antivirais/química , Antivirais/farmacocinética , Antivirais/farmacologia , Biologia Computacional/métodos , Desenho de Fármacos , Humanos , Peptídeos/farmacologia , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/efeitos dos fármacos , Proteínas Virais de Fusão/genética
12.
Vopr Virusol ; 53(3): 12-5, 2008.
Artigo em Russo | MEDLINE | ID: mdl-18590128

RESUMO

A model of peritoneal macrophages was used to study the effect of fucoidans from brown seaweeds on the adsorption of Hantaan virus. Fucoidans were found to have antiviral activity, but to differ in the inhibition of hantavirus adsorption, which was associated with their structural features. The mechanism of their action is assumed to be shown via ligand-receptor interaction with certain cell membrane receptors and via blockage of hantavirus glycoproteins (G1 and G2), resulting in adsorption inhibition and preventing viral penetration into the macrophages.


Assuntos
Vírus Hantaan/efeitos dos fármacos , Macrófagos Peritoneais/virologia , Polissacarídeos/farmacologia , Animais , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Vírus Hantaan/fisiologia , Febre Hemorrágica com Síndrome Renal/tratamento farmacológico , Febre Hemorrágica com Síndrome Renal/virologia , Camundongos , Células Vero , Proteínas Virais de Fusão/efeitos dos fármacos , Ligação Viral/efeitos dos fármacos
13.
Nat Immunol ; 6(10): 995-1001, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16155572

RESUMO

Defensins are peptides that protect the host against microorganisms. Here we show that the theta-defensin retrocyclin 2 (RC2) inhibited influenza virus infection by blocking membrane fusion mediated by the viral hemagglutinin. RC2 was effective even after hemagglutinin attained a fusogenic conformation or had induced membrane hemifusion. RC2, a multivalent lectin, prevented hemagglutinin-mediated fusion by erecting a network of crosslinked and immobilized surface glycoproteins. RC2 also inhibited fusion mediated by Sindbis virus and baculovirus. Human beta-defensin 3 and mannan-binding lectin also blocked viral fusion by creating a protective barricade of immobilized surface proteins. This general mechanism might explain the broad-spectrum antiviral activity of many multivalent lectins of the innate immune system.


Assuntos
Anti-Infecciosos/farmacologia , Metabolismo dos Carboidratos , Defensinas/farmacologia , Glicoproteínas de Membrana/efeitos dos fármacos , Infecções por Orthomyxoviridae/imunologia , Orthomyxoviridae/fisiologia , Linhagem Celular Tumoral , Eritrócitos , Hemaglutininas Virais/metabolismo , Humanos , Imunidade Inata , Fusão de Membrana/efeitos dos fármacos , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/metabolismo , Orthomyxoviridae/efeitos dos fármacos , Infecções por Orthomyxoviridae/virologia , Proteínas do Envelope Viral/efeitos dos fármacos , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/efeitos dos fármacos , Proteínas Virais de Fusão/metabolismo , Replicação Viral/efeitos dos fármacos
15.
Antimicrob Agents Chemother ; 48(5): 1495-502, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15105096

RESUMO

Human parainfluenza viruses are important respiratory tract pathogens, especially of children. However, no vaccines or specific therapies for infections caused by these viruses are currently available. In the present study we characterized the efficacy of the novel parainfluenza virus inhibitors BCX 2798 and BCX 2855, which were designed based on the three-dimensional structure of the hemagglutinin-neuraminidase (HN) protein. The compounds were highly effective in inhibiting hemagglutinin (HA) and neuraminidase (NA) activities and the growth of hPIV-1, hPIV-2, and hPIV-3 in LLC-MK(2) cells. The concentrations required to reduce the activity to 50% of that of a control ranged from 0.1 to 6.0 micro M in HA inhibition assays and from 0.02 to 20 micro M in NA inhibition assays. The concentrations required to inhibit virus replication to 50% of the level of the control ranged from 0.7 to 11.5 micro M. BCX 2798 and BCX 2855 were inactive against influenza virus HA and NA and bacterial NA. In mice infected with a recombinant Sendai virus whose HN gene was replaced with that of hPIV-1 [rSV(hHN)], intranasal administration of BCX 2798 (10 mg/kg per day) and of BCX 2855 (50 mg/kg per day) 4 h before the start of infection resulted in a significant reduction in titers of virus in the lungs and protection from death. Treatment beginning 24 h after the start of infection did not prevent death. Together, our results indicate that BCX 2798 and BCX 2855 are effective inhibitors of parainfluenza virus HN and may limit parainfluenza virus infections in humans.


Assuntos
Antivirais/farmacologia , Azidas/farmacologia , Inibidores Enzimáticos/farmacologia , Hemaglutininas/efeitos dos fármacos , Ácidos Hexurônicos/farmacologia , Neuraminidase/antagonistas & inibidores , Vírus da Parainfluenza 1 Humana/efeitos dos fármacos , Vírus da Parainfluenza 2 Humana/efeitos dos fármacos , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Sulfonamidas/farmacologia , Animais , Feminino , Humanos , Células LLC-PK1 , Pulmão/patologia , Pulmão/virologia , Camundongos , Infecções por Paramyxoviridae/tratamento farmacológico , Infecções por Paramyxoviridae/patologia , Infecções por Paramyxoviridae/virologia , Proteínas Recombinantes , Vírus Sendai/efeitos dos fármacos , Vírus Sendai/genética , Suínos , Proteínas Virais de Fusão/efeitos dos fármacos , Proteínas Virais de Fusão/genética , Replicação Viral/efeitos dos fármacos
16.
EMBO J ; 21(17): 4402-10, 2002 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-12198142

RESUMO

In the icosahedral (T = 4) Semliki Forest virus, the envelope protomers, i.e. E1-E2 heterodimers, make one-to-one interactions with capsid proteins below the viral lipid bilayer, transverse the membrane and form an external glycoprotein shell with projections. The shell is organized by protomer domains interacting as hexamers and pentamers around shell openings at icosahedral 2- and 5-fold axes, respectively, and the projections by other domains associating as trimers at 3- and quasi 3-fold axes. We show here, using cryo- electron microscopy, that low pH, as occurs in the endosomes during virus uptake, results in the relaxation of protomer interactions around the 2- and the 5-fold axes in the shell, and movement of protomers towards 3- and quasi 3-fold axes in a way that reciprocally relocates their putative E1 and E2 domains. This seemed to be facilitated by a trimerization of transmembrane segments at the same axes. The alterations observed help to explain several key features of the spike-mediated membrane fusion reaction, including shell dissolution, heterodimer dissociation, fusion peptide exposure and E1 homotrimerization.


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Fusão de Membrana/efeitos dos fármacos , Morfolinas/farmacologia , Vírus da Floresta de Semliki/química , Proteínas Virais de Fusão/ultraestrutura , Animais , Capsídeo/efeitos dos fármacos , Capsídeo/ultraestrutura , Linhagem Celular/virologia , Cricetinae , Microscopia Crioeletrônica , Efeito Citopatogênico Viral , Dimerização , Endossomos/virologia , Concentração de Íons de Hidrogênio , Processamento de Imagem Assistida por Computador , Rim , Bicamadas Lipídicas , Mesocricetus , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/efeitos dos fármacos , Proteínas Virais de Fusão/fisiologia
17.
J Virol ; 75(11): 5230-9, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11333905

RESUMO

A CD4-independent version of the X4 human immunodeficiency virus type 1 (HIV-1) HXBc2 envelope (Env) protein, termed 8x, mediates infection of CD4-negative, CXCR4-positive cells, binds directly to CXCR4 in the absence of CD4 due to constitutive exposure of a conserved coreceptor binding site in the gp120 subunit, and is more sensitive to antibody-mediated neutralization. To study the relationships between CD4 independence, neutralization sensitivity, and exposure of CD4-induced epitopes associated with the coreceptor binding site, we generated a large panel of Env mutants and chimeras between 8x and its CD4-dependent parent, HXBc2. We found that a frameshift mutation just proximal to the gp41 cytoplasmic domain in 8x Env was necessary but not sufficient for CD4 independence and led to increased exposure of the coreceptor binding site. In the presence of this altered cytoplasmic domain, single amino acid changes in either the 8x V3 (V320I) or V4/C4 (N386K) regions imparted CD4 independence, with other changes playing a modulatory role. The N386K mutation resulted in loss of an N-linked glycosylation site, but additional mutagenesis showed that it was the presence of a lysine rather than loss of the glycosylation site that contributed to CD4 independence. However, loss of the glycosylation site alone was sufficient to render Env neutralization sensitive, providing additional evidence that carbohydrate structures shield important neutralization determinants. Exposure of the CD4-induced epitope recognized by monoclonal antibody 17b and which overlaps the coreceptor binding site was highly sensitive to an R298K mutation at the base of the V3 loop and was often but not always associated with CD4 independence. Finally, while not all neutralization-sensitive Envs were CD4 independent, all CD4-independent Envs exhibited enhanced sensitivity to neutralization by HIV-1-positive human sera, indicating that the humoral immune response can exert strong selective pressure against the CD4-independent phenotype in vivo. Whether this can be used to advantage in designing more effective immunogens remains to be seen.


Assuntos
Antígenos CD4/metabolismo , Proteína gp120 do Envelope de HIV/metabolismo , Proteína gp41 do Envelope de HIV/metabolismo , HIV-1/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Sítios de Ligação , Proteínas Sanguíneas/farmacologia , Antígenos CD4/genética , Fusão Celular , Linhagem Celular , Epitopos/metabolismo , Glicosilação , Proteína gp120 do Envelope de HIV/genética , Proteína gp41 do Envelope de HIV/genética , HIV-1/metabolismo , Humanos , Mutagênese Sítio-Dirigida , Testes de Neutralização , Conformação Proteica , Codorniz , Receptores CXCR4/metabolismo , Transfecção , Proteínas Virais de Fusão/efeitos dos fármacos
18.
Biosci Rep ; 21(3): 293-304, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11892996

RESUMO

We have directly compared the effect of two types of dextran sulfate with distinct molecular weights (500 kDa and 5 kDa) on the fusion activity and infectivity of both Sendai and influenza viruses, two lipid-enveloped viruses that differ in their routes of entry into target cells. To correlate membrane merging and infectivity MDCK cells were used as targets for the viruses in both approaches. In either case pronounced inhibition of virus-cell interactions by dextran sulfate was only observed at low pH, even though Sendai virus fuses maximally at pH 7.4. Although membrane merging could not be fully abolished, the inhibitory effect was always greater when the higher molecular weight dextran sulfate was used. The presence of this residual fusion activity, that could not be reduced even with high concentrations of agent, suggests that a limited number of binding sites for dextran sulfate may exist on the viral envelopes. The compounds also inhibited fusion of bound virions, and all results could be reproduced using erythrocyte ghosts as target membranes in the fusion assay, instead of MDCK cells. In agreement with these observations only the infectivity of influenza virus (which requires a low pH-dependent step to enter target cells) was affected by dextran sulfate, again the higher molecular weight compound showing a more pronounced inhibitory effect.


Assuntos
Sulfato de Dextrana/farmacologia , Influenza Humana/tratamento farmacológico , Fusão de Membrana/efeitos dos fármacos , Orthomyxoviridae/efeitos dos fármacos , Infecções por Respirovirus/tratamento farmacológico , Vírus Sendai/efeitos dos fármacos , Proteínas Virais de Fusão/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Membrana Celular/virologia , Células Cultivadas , Endocitose/efeitos dos fármacos , Endocitose/imunologia , Membrana Eritrocítica/efeitos dos fármacos , Membrana Eritrocítica/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Influenza Humana/metabolismo , Influenza Humana/fisiopatologia , Fusão de Membrana/fisiologia , Peso Molecular , Orthomyxoviridae/metabolismo , Orthomyxoviridae/patogenicidade , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Infecções por Respirovirus/metabolismo , Infecções por Respirovirus/fisiopatologia , Vírus Sendai/metabolismo , Vírus Sendai/patogenicidade , Proteínas Virais de Fusão/metabolismo
19.
Bioorg Khim ; 24(9): 676-81, 1998 Sep.
Artigo em Russo | MEDLINE | ID: mdl-9813732

RESUMO

The synthetic peptide with the conservative 98-113 sequence of protein E of tick-borne encephalitis virus was studied in order to elucidate its role in the functioning of flaviviruses. The peptide was shown to inhibit the in vitro infection of macrophages with the virus. An antibody that specifically binds this peptide was found among the set of monoclonal antibodies produced against protein E. This antibody was found to prevent penetration of the virus into liposomes. A correlation was found between our results and data on the spatial structure of protein E and its interspecies homology. The protein E 98-113 sequence of the tick-borne encephalitis virus was found to be the fusion site of the viral envelope with a cellular membrane.


Assuntos
Antígenos Virais/fisiologia , Vírus da Encefalite Transmitidos por Carrapatos/fisiologia , Fragmentos de Peptídeos/farmacologia , Proteínas do Envelope Viral/fisiologia , Proteínas Virais de Fusão/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Antígenos Virais/química , Antígenos Virais/imunologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/virologia , Relação Dose-Resposta a Droga , Técnicas In Vitro , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/virologia , Camundongos , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Baço/citologia , Baço/efeitos dos fármacos , Baço/virologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/imunologia
20.
Virology ; 238(2): 291-304, 1997 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-9400602

RESUMO

To investigate the molecular mechanisms involved in paramyxovirus-induced cell fusion, the function and structure of a peptide with a 20-amino-acid sequence from the leucine zipper region (heptad repeat region 2) of the Newcastle disease virus fusion protein (F) were characterized. A peptide with the sequence ALDKLEESNSKLDKVNVKLT (amino acids 478-497 of the F protein) was found to inhibit syncytia formation after virus infection and after transfection of Cos cells with the HN (hemagglutinin-neuraminidase) and F protein cDNAs. Using an F protein gene that requires addition of exogenous trypsin for cleavage, it was shown that the peptide exerted its inhibitory effect prior to cleavage. The three-dimensional conformation of the peptide in aqueous solution was determined through the use of NMR and molecular modeling. Results showed that the peptide formed a helix with properties between an alpha-helix and a 3(10)-helix and that leucine residues aligned along one face of the helix. Side chain salt bridges and hydrogen bonds likely contributed to the stability of the peptide secondary structure. Analysis of the aqueous solution conformation of the peptide suggested mechanisms for specificity of interaction with the intact F protein.


Assuntos
Fusão de Membrana/efeitos dos fármacos , Vírus da Doença de Newcastle/efeitos dos fármacos , Peptídeos/farmacologia , Proteínas Virais de Fusão/efeitos dos fármacos , Amidas/química , Sequência de Aminoácidos , Animais , Células COS , Proteína HN/genética , Proteína HN/metabolismo , Zíper de Leucina , Modelos Moleculares , Dados de Sequência Molecular , Vírus da Doença de Newcastle/fisiologia , Ressonância Magnética Nuclear Biomolecular , Biossíntese Peptídica , Fragmentos de Peptídeos , Peptídeos/química , Estrutura Secundária de Proteína , Temperatura , Fatores de Tempo , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...