Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
mBio ; 11(1)2020 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-32047132

RESUMO

The receptor binding protein of parainfluenza virus, hemagglutinin-neuraminidase (HN), is responsible for actively triggering the viral fusion protein (F) to undergo a conformational change leading to insertion into the target cell and fusion of the virus with the target cell membrane. For proper viral entry to occur, this process must occur when HN is engaged with host cell receptors at the cell surface. It is possible to interfere with this process through premature activation of the F protein, distant from the target cell receptor. Conformational changes in the F protein and adoption of the postfusion form of the protein prior to receptor engagement of HN at the host cell membrane inactivate the virus. We previously identified small molecules that interact with HN and induce it to activate F in an untimely fashion, validating a new antiviral strategy. To obtain highly active pretriggering candidate molecules we carried out a virtual modeling screen for molecules that interact with sialic acid binding site II on HN, which we propose to be the site responsible for activating F. To directly assess the mechanism of action of one such highly effective new premature activating compound, PAC-3066, we use cryo-electron tomography on authentic intact viral particles for the first time to examine the effects of PAC-3066 treatment on the conformation of the viral F protein. We present the first direct observation of the conformational rearrangement induced in the viral F protein.IMPORTANCE Paramyxoviruses, including human parainfluenza virus type 3, are internalized into host cells by fusion between viral and target cell membranes. The receptor binding protein, hemagglutinin-neuraminidase (HN), upon binding to its cell receptor, triggers conformational changes in the fusion protein (F). This action of HN activates F to reach its fusion-competent state. Using small molecules that interact with HN, we can induce the premature activation of F and inactivate the virus. To obtain highly active pretriggering compounds, we carried out a virtual modeling screen for molecules that interact with a sialic acid binding site on HN that we propose to be the site involved in activating F. We use cryo-electron tomography of authentic intact viral particles for the first time to directly assess the mechanism of action of this treatment on the conformation of the viral F protein and present the first direct observation of the induced conformational rearrangement in the viral F protein.


Assuntos
Antivirais/farmacologia , Proteína HN/metabolismo , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Proteínas Virais de Fusão/antagonistas & inibidores , Internalização do Vírus/efeitos dos fármacos , Antivirais/isolamento & purificação , Técnicas de Cultura de Células , Linhagem Celular , Descoberta de Drogas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Proteína HN/genética , Ensaios de Triagem em Larga Escala , Humanos , Simulação de Acoplamento Molecular , Vírus da Parainfluenza 3 Humana/fisiologia , Infecções por Paramyxoviridae/tratamento farmacológico , Ligação Proteica/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas Virais de Fusão/metabolismo
2.
mBio ; 10(1)2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30782664

RESUMO

Paramyxoviruses, specifically, the childhood pathogen human parainfluenza virus type 3, are internalized into host cells following fusion between the viral and target cell membranes. The receptor binding protein, hemagglutinin (HA)-neuraminidase (HN), and the fusion protein (F) facilitate viral fusion and entry into the cell through a coordinated process involving HN activation by receptor binding, which triggers conformational changes in the F protein to activate it to reach its fusion-competent state. Interfering with this process through premature activation of the F protein has been shown to be an effective antiviral strategy in vitro. Conformational changes in the F protein leading to adoption of the postfusion form of the protein-prior to receptor engagement of HN at the host cell membrane-render the virus noninfectious. We previously identified a small compound (CSC11) that implements this antiviral strategy through an interaction with HN, causing HN to activate F in an untimely process. To assess the functionality of such compounds, it is necessary to verify that the postfusion state of F has been achieved. As demonstrated by Melero and colleagues, soluble forms of the recombinant postfusion pneumovirus F proteins and of their six helix bundle (6HB) motifs can be used to generate postfusion-specific antibodies. We produced novel anti-HPIV3 F conformation-specific antibodies that can be used to assess the functionality of compounds designed to induce F activation. In this study, using systematic chemical modifications of CSC11, we synthesized a more potent derivative of this compound, CM9. Much like CSC11, CM9 causes premature triggering of the F protein through an interaction with HN prior to receptor engagement, thereby preventing fusion and subsequent infection. In addition to validating the potency of CM9 using plaque reduction, fusion inhibition, and binding avidity assays, we confirmed the transition to a postfusion conformation of F in the presence of CM9 using our novel anti-HPIV3 conformation-specific antibodies. We present both CM9 and these newly characterized postfusion antibodies as novel tools to explore and develop antiviral approaches. In turn, these advances in both our molecular toolset and our understanding of HN-F interaction will support development of more-effective antivirals. Combining the findings described here with our recently described physiologically relevant ex vivo system, we have the potential to inform the development of therapeutics to block viral infection.IMPORTANCE Paramyxoviruses, including human parainfluenza virus type 3, are internalized into host cells by fusion between viral and target cell membranes. The receptor binding protein, hemagglutinin-neuraminidase (HN), and the fusion protein (F) facilitate viral fusion and entry into cells through a process involving HN activation by receptor binding, which triggers conformational changes in F to activate it to reach its fusion-competent state. Interfering with this process through premature activation of the F protein may be an effective antiviral strategy in vitro We identified and optimized small compounds that implement this antiviral strategy through an interaction with HN, causing HN to activate F in an untimely fashion. To address that mechanism, we produced novel anti-HPIV3 F conformation-specific antibodies that can be used to assess the functionality of compounds designed to induce F activation. Both the novel antiviral compounds that we present and these newly characterized postfusion antibodies are novel tools for the exploration and development of antiviral approaches.


Assuntos
Antivirais/farmacologia , Proteína HN/metabolismo , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Vírus da Parainfluenza 3 Humana/fisiologia , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus/efeitos dos fármacos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/isolamento & purificação , Antivirais/síntese química , Linhagem Celular , Chlorocebus aethiops , Humanos , Ligação Proteica , Conformação Proteica , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/imunologia , Ensaio de Placa Viral
3.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27733647

RESUMO

Measles virus (MV) infection is undergoing resurgence and remains one of the leading causes of death among young children worldwide despite the availability of an effective measles vaccine. MV infects its target cells by coordinated action of the MV hemagglutinin (H) and fusion (F) envelope glycoproteins; upon receptor engagement by H, the prefusion F undergoes a structural transition, extending and inserting into the target cell membrane and then refolding into a postfusion structure that fuses the viral and cell membranes. By interfering with this structural transition of F, peptides derived from the heptad repeat (HR) regions of F can inhibit MV infection at the entry stage. In previous work, we have generated potent MV fusion inhibitors by dimerizing the F-derived peptides and conjugating them to cholesterol. We have shown that prophylactic intranasal administration of our lead fusion inhibitor efficiently protects from MV infection in vivo We show here that peptides tagged with lipophilic moieties self-assemble into nanoparticles until they reach the target cells, where they are integrated into cell membranes. The self-assembly feature enhances biodistribution and the half-life of the peptides, while integration into the target cell membrane increases fusion inhibitor potency. These factors together modulate in vivo efficacy. The results suggest a new framework for developing effective fusion inhibitory peptides. IMPORTANCE: Measles virus (MV) infection causes an acute illness that may be associated with infection of the central nervous system (CNS) and severe neurological disease. No specific treatment is available. We have shown that fusion-inhibitory peptides delivered intranasally provide effective prophylaxis against MV infection. We show here that specific biophysical properties regulate the in vivo efficacy of MV F-derived peptides.


Assuntos
Hemaglutininas Virais/imunologia , Vacina contra Sarampo/administração & dosagem , Vírus do Sarampo/efeitos dos fármacos , Sarampo/prevenção & controle , Nanopartículas/administração & dosagem , Peptídeos/imunologia , Proteínas Virais de Fusão/imunologia , Administração Intranasal , Sequência de Aminoácidos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Colesterol/química , Feminino , Meia-Vida , Hemaglutininas Virais/química , Humanos , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Masculino , Sarampo/imunologia , Sarampo/mortalidade , Sarampo/virologia , Vacina contra Sarampo/síntese química , Vírus do Sarampo/química , Vírus do Sarampo/imunologia , Nanopartículas/química , Peptídeos/síntese química , Sigmodontinae , Análise de Sobrevida , Proteínas Virais de Fusão/química , Internalização do Vírus/efeitos dos fármacos
4.
J Virol ; 89(2): 1143-55, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25378493

RESUMO

UNLABELLED: Measles virus (MV) infection is undergoing resurgence and remains one of the leading causes of death among young children worldwide despite the availability of an effective measles vaccine. MV infects its target cells by coordinated action of the MV H and the fusion (F) envelope glycoprotein; upon receptor engagement by H, the prefusion F undergoes a structural transition, extending and inserting into the target cell membrane and then refolding into a postfusion structure that fuses the viral and cell membranes. By interfering with this structural transition of F, peptides derived from the heptad-repeat (HR) regions of F can potently inhibit MV infection at the entry stage. We show here that specific features of H's interaction with its receptors modulate the susceptibility of MV F to peptide fusion inhibitors. A higher concentration of inhibitory peptides is required to inhibit F-mediated fusion when H is engaged to its nectin-4 receptor than when H is engaged to its CD150 receptor. Peptide inhibition of F may be subverted by continued engagement of receptor by H, a finding that highlights the ongoing role of H-receptor interaction after F has been activated and that helps guide the design of more potent inhibitory peptides. Intranasal administration of these peptides results in peptide accumulation in the airway epithelium with minimal systemic levels of peptide and efficiently prevents MV infection in vivo in animal models. The results suggest an antiviral strategy for prophylaxis in vulnerable and/or immunocompromised hosts. IMPORTANCE: Measles virus (MV) infection causes an acute illness that may be associated with infection of the central nervous system (CNS) and severe neurological disease. No specific treatment is available. We have shown that parenterally delivered fusion-inhibitory peptides protect mice from lethal CNS MV disease. Here we show, using established small-animal models of MV infection, that fusion-inhibitory peptides delivered intranasally provide effective prophylaxis against MV infection. Since the fusion inhibitors are stable at room temperature, this intranasal strategy is feasible even outside health care settings, could be used to protect individuals and communities in case of MV outbreaks, and could complement global efforts to control measles.


Assuntos
Antivirais/administração & dosagem , Vírus do Sarampo/efeitos dos fármacos , Sarampo/prevenção & controle , Oligopeptídeos/administração & dosagem , Proteínas Virais de Fusão/administração & dosagem , Internalização do Vírus/efeitos dos fármacos , Administração Intranasal , Animais , Quimioprevenção/métodos , Modelos Animais de Doenças , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Sigmodontinae
5.
J Virol ; 81(19): 10567-74, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17652384

RESUMO

Hendra virus (HeV) and Nipah virus (NiV) constitute the Henipavirus genus of paramyxoviruses, both fatal in humans and with the potential for subversion as agents of bioterrorism. Binding of the HeV/NiV attachment protein (G) to its receptor triggers a series of conformational changes in the fusion protein (F), ultimately leading to formation of a postfusion six-helix bundle (6HB) structure and fusion of the viral and cellular membranes. The ectodomain of paramyxovirus F proteins contains two conserved heptad repeat regions, the first (the N-terminal heptad repeat [HRN]) adjacent to the fusion peptide and the second (the C-terminal heptad repeat [HRC]) immediately preceding the transmembrane domain. Peptides derived from the HRN and HRC regions of F are proposed to inhibit fusion by preventing activated F molecules from forming the 6HB structure that is required for fusion. We previously reported that a human parainfluenza virus 3 (HPIV3) F peptide effectively inhibits infection mediated by the HeV glycoproteins in pseudotyped-HeV entry assays more effectively than the comparable HeV-derived peptide, and we now show that this peptide inhibits live-HeV and -NiV infection. HPIV3 F peptides were also effective in inhibiting HeV pseudotype virus entry in a new assay that mimics multicycle replication. This anti-HeV/NiV efficacy can be correlated with the greater potential of the HPIV3 C peptide to interact with the HeV F N peptide coiled-coil trimer, as evaluated by thermal unfolding experiments. Furthermore, replacement of a buried glutamic acid (glutamic acid 459) in the C peptide with valine enhances antiviral potency and stabilizes the 6HB conformation. Our results strongly suggest that conserved interhelical packing interactions in the F protein fusion core are important determinants of C peptide inhibitory activity and offer a strategy for the development of more-potent analogs of F peptide inhibitors.


Assuntos
Antivirais/farmacologia , Henipavirus/efeitos dos fármacos , Mimetismo Molecular , Peptídeos/farmacologia , Fosfoproteínas/farmacologia , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas Virais/farmacologia , Internalização do Vírus/efeitos dos fármacos , Sequência de Aminoácidos , Antivirais/química , Linhagem Celular , Sequência Conservada , Vírus Hendra/efeitos dos fármacos , Vírus Hendra/fisiologia , Henipavirus/fisiologia , Humanos , Dados de Sequência Molecular , Mutação , Vírus Nipah/efeitos dos fármacos , Vírus Nipah/fisiologia , Paramyxovirinae/efeitos dos fármacos , Peptídeos/química , Peptídeos/genética , Fosfoproteínas/química , Fosfoproteínas/genética , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas Virais/química , Proteínas Virais/genética
6.
J Virol ; 80(19): 9837-49, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16973588

RESUMO

Hendra virus (HeV) is a recently identified paramyxovirus that is fatal in humans and could be used as an agent of bioterrorism. The HeV receptor-binding protein (G) is required in order for the fusion protein (F) to mediate fusion, and analysis of the triggering/activation of HeV F by G should lead to strategies for interfering with this key step in viral entry. HeV F, once triggered by the receptor-bound G, by analogy with other paramyxovirus F proteins, undergoes multistep conformational changes leading to a six-helix bundle (6HB) structure that accomplishes fusion of the viral and cellular membranes. The ectodomain of paramyxovirus F proteins contains two conserved heptad repeat regions (HRN and HRC) near the fusion peptide and the transmembrane domains, respectively. Peptides derived from the HRN and HRC regions of F are proposed to inhibit fusion by preventing F, after the initial triggering step, from forming the 6HB structure that is required for fusion. HeV peptides have previously been found to be effective at inhibiting HeV fusion. However, we found that a human parainfluenza virus 3 F-peptide is more effective at inhibiting HeV fusion than the comparable HeV-derived peptide.


Assuntos
Vírus Hendra/fisiologia , Sequência de Aminoácidos , Fusão Celular , Linhagem Celular , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutação/genética , Fragmentos de Peptídeos/metabolismo , Estrutura Terciária de Proteína , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo , Vírion/metabolismo
7.
J Pediatr Gastroenterol Nutr ; 33(4): 445-9, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11698761

RESUMO

BACKGROUND: Risk factors for the development of posttransplant lymphoproliferative disease (PTLD), a major cause of morbidity and mortality after pediatric liver transplantation, are primary Epstein-Barr virus (EBV) infection and intensity of immunosuppression. The authors assessed monitoring of EBV replication and preemptive immunosuppression reduction in pediatric liver transplant recipients. METHODS: The authors prospectively followed monthly EBV-quantitative competitive polymerase chain reaction to measure EBV replication in 23 patients who underwent liver transplant between July 1997 and November 1998. Preemptive immunosuppression reduction was instituted for significant EBV replication. Patients were followed up for at least 1 year and divided in two groups for analysis (group 1, pretransplant seronegative for EBV [13 patients]; group 2, seropositive for EBV [10 patients]). RESULTS: In group 1, 9 of 13 patients had positive polymerase chain reaction results at a mean time of 22.4 weeks after transplantation. All but one of these patients were asymptomatic. In seven of nine patients, preemptive immunosuppression reduction was undertaken without development of PTLD or rejection. In two of nine patients, immunosuppression could not be continuously reduced, and both patients experienced low-grade and medically responsive PTLD. In no patient in group 2 did an EBV-positive viral load or PTLD develop. CONCLUSIONS: Prospective longitudinal measurement of EBV by quantitative competitive polymerase chain reaction permits early detection of asymptomatic viral replication. Subsequent preemptive reduction of immunosuppression may prevent the progression to PTLD.


Assuntos
Infecções por Vírus Epstein-Barr/complicações , Transtornos Linfoproliferativos/etiologia , Transtornos Linfoproliferativos/prevenção & controle , Transplante de Órgãos/efeitos adversos , Complicações Pós-Operatórias/etiologia , Adolescente , Adulto , Criança , Pré-Escolar , Estudos de Coortes , Infecções por Vírus Epstein-Barr/transmissão , Evolução Fatal , Feminino , Rejeição de Enxerto/prevenção & controle , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/isolamento & purificação , Humanos , Terapia de Imunossupressão/efeitos adversos , Imunossupressores/administração & dosagem , Lactente , Recém-Nascido , Transtornos Linfoproliferativos/virologia , Masculino , Reação em Cadeia da Polimerase , Complicações Pós-Operatórias/virologia , Estudos Prospectivos , Fatores de Risco , Tacrolimo/administração & dosagem , Carga Viral
8.
Virology ; 283(2): 230-9, 2001 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-11336548

RESUMO

Type I interferon (IFN) induces antiviral responses through the activation of the ISGF3 transcription factor complex that contains the subunit proteins STAT1, STAT2, and p48/ISGF3 gamma/IRF9. The ability of some human paramyxoviruses to overcome IFN actions by specific proteolysis of STAT proteins has been examined. Infection of cells with type 2, but not type 1 or type 3 human parainfluenza virus (HPIV) leads to a loss of cellular STAT2 protein. Expression of a single HPIV2 protein derived from the V open reading frame blocks IFN-dependent transcriptional responses in the absence of other viral proteins. The loss of IFN response is due to V-protein-induced proteolytic degradation of STAT2. Expression of HPIV2 V causes the normally stable STAT2 protein to be rapidly degraded, and this proteolytic activity can be partially alleviated by proteasome inhibition. No V-protein-specific effects on STAT2 mRNA levels were observed. The results indicate that the V protein of HPIV2 is sufficient to recognize and target a specific cellular transcription factor for destruction by cellular machinery.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Interferon Tipo I/imunologia , Vírus da Parainfluenza 2 Humana/patogenicidade , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Virais , Proteínas Estruturais Virais/metabolismo , Linhagem Celular , Cisteína Endopeptidases/metabolismo , DNA Complementar , Humanos , Fator Gênico 3 Estimulado por Interferon , Fator Gênico 3 Estimulado por Interferon, Subunidade gama , Complexos Multienzimáticos/antagonistas & inibidores , Complexos Multienzimáticos/metabolismo , Vírus da Parainfluenza 2 Humana/genética , Vírus da Parainfluenza 2 Humana/metabolismo , Complexo de Endopeptidases do Proteassoma , Processamento de Proteína Pós-Traducional , RNA Mensageiro/metabolismo , Infecções por Rubulavirus/fisiopatologia , Infecções por Rubulavirus/virologia , Fator de Transcrição STAT1 , Fator de Transcrição STAT2 , Transfecção , Proteínas Estruturais Virais/genética
9.
Transplantation ; 67(7): 1068-70, 1999 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10221496

RESUMO

BACKGROUND: Posttransplant lymphoproliferative disease (PTLD) remains a significant cause of morbidity and mortality in pediatric liver transplant recipients. Epstein-Barr Virus (EBV) mismatch associated with more prevalent use of split-liver, reduced size, and living-related transplants has increased the risk of primary EBV infection and subsequent PTLD. Early identification of EBV viremia may reduce the risk of PTLD, because it allows for early adjustment of immunosuppression and antiviral therapy. METHODS: EBV viral load was measured monthly by quantitative competitive polymerase chain reactions in three pediatric liver transplant recipients. RESULTS: Onset of EBV viremia was documented in one recipient. Established EBV viremia was followed in the other two recipients (one with chronic rejection and one with PTLD) who were initially tested once monitoring was initiated in our program. CONCLUSIONS: EBV quantitative competitive polymerase chain reactions may represent a promising way to follow EBV viral load and potentially prevent the development of PTLD.


Assuntos
Herpesvirus Humano 4/isolamento & purificação , Transplante de Fígado , Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/etiologia , Feminino , Rejeição de Enxerto/complicações , Humanos , Lactente , Estudos Longitudinais , Transtornos Linfoproliferativos/complicações , Transtornos Linfoproliferativos/etiologia , Masculino , Reação em Cadeia da Polimerase , Complicações Pós-Operatórias , Período Pós-Operatório , Estudos Prospectivos , Carga Viral , Viremia/complicações , Viremia/etiologia
11.
Microb Pathog ; 20(3): 179-84, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8965678

RESUMO

In order for the fusion protein (F) of the human parainfluenza virus type 3 (HPF3) to promote membrane fusion and viral entry, the haemagglutinin-neuraminidase (HN) glycoprotein must interact with its receptor. Sialoglycoconjugates are known to be the receptors for the HPF3 HN, however specific attachment factors or receptors for HPF3 have not been identified. In this report we describe the analysis of variants of HPF3 with increased fusion-promoting phenotypes that were selected by treatment with viral neuraminidase. The results suggest that for HPF3, the virus is specific in its use of sialic acid receptors; the majority of sialic-acid containing molecules are not targets for HPF3.


Assuntos
Vírus da Parainfluenza 3 Humana/química , Receptores Virais/análise , Variação Genética , Neuraminidase/metabolismo , Proteínas Virais de Fusão/análise
12.
J Virol ; 67(11): 6463-8, 1993 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8411349

RESUMO

The ability of enveloped viruses to cause disease depends on their ability to enter the host cell via membrane fusion events. An understanding of these early events in infection, crucial for the design of methods of blocking infection, is needed for viruses that mediate membrane fusion at neutral pH, such as paramyxoviruses and human immunodeficiency virus. Sialic acid is the receptor for the human parainfluenza virus type 3 (HPF3) hemagglutinin-neuraminidase (HN) glycoprotein, the molecule responsible for binding of the virus to cell surfaces. In order for the fusion protein (F) of HPF3 to promote membrane fusion, the HN must interact with its receptor. In the present report, two variants of HPF3 with increased fusion-promoting phenotypes were selected and used to study the function of the HN glycoprotein in membrane fusion. Increased fusogenicity correlated with single amino acid changes in the HN protein that resulted in increased binding of the variant viruses to the sialic acid receptor. These results suggest that the avidity of binding of the HN protein to its receptor regulates the level of F protein-mediated fusion and begin to define one role of the receptor-binding protein of a paramyxovirus in the membrane fusion process.


Assuntos
Fusão Celular , Proteína HN/metabolismo , Vírus da Parainfluenza 3 Humana/patogenicidade , Receptores Virais/metabolismo , Ácidos Siálicos/metabolismo , Proteínas Virais de Fusão/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Técnicas In Vitro
13.
J Virol ; 66(11): 6280-7, 1992 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-1328668

RESUMO

Cells can be persistently infected with human parainfluenza virus type 3 (HPF3) by using a high multiplicity of infection (MOI) (> or = 5 PFU per cell). The persistently infected cells exhibit no cytopathic effects and do not fuse with each other, yet they readily fuse with uninfected cells. We have previously shown that the failure of the persistently infected cells to fuse with each other is due to the lack of a receptor on these cells for the viral hemagglutinin-neuraminidase glycoprotein, and we have established that both fusion and hemagglutinin-neuraminidase proteins are needed for cell fusion mediated by HPF3. We then postulated that the generation of persistent infection and the failure of cells infected with HPF3 at high MOI to form syncytia are both due to the action of viral neuraminidase in the high-MOI inoculum. In this report, we describe experiments to test this hypothesis and further investigate the receptor requirements for HPF3 infection and cell fusion. A normally cytopathic low-MOI HPF3 infection can be converted into a noncytopathic infection by the addition of exogenous neuraminidase, either in the form of a purified enzyme or as UV-inactivated HPF3 virions. Evidence is presented that the receptor requirements for an HPF3 virus particle to infect a cell are different from those for fusion between cells. By treating infected cells in culture with various doses of neuraminidase, we demonstrate that virus spreads from cell to cell in the complete absence of cell-cell fusion. We compare the outcome of HPF3 infection in the presence of excess neuraminidase with that of another paramyxovirus (simian virus 5) and provide evidence that these two viruses differ in their receptor requirements for mediating fusion.


Assuntos
Fusão Celular , Fusão de Membrana , Vírus da Parainfluenza 3 Humana/crescimento & desenvolvimento , Receptores Virais/metabolismo , Células Cultivadas , Hemaglutininas Virais/metabolismo , Humanos , Modelos Biológicos , Ácido N-Acetilneuramínico , Neuraminidase/metabolismo , Vírus da Parainfluenza 3 Humana/patogenicidade , Retrovirus dos Símios/metabolismo , Retrovirus dos Símios/patogenicidade , Ácidos Siálicos/metabolismo , Proteínas Virais de Fusão/metabolismo , Replicação Viral
14.
J Am Acad Dermatol ; 26(3 Pt 2): 462-6, 1992 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-1564154

RESUMO

BACKGROUND: Malignant melanomas (MMs) with a small diameter (less than 6 mm) are rarely removed surgically; therefore there is a need for more data to characterize them. OBJECTIVE: The purpose of this study was to further elucidate the clinical and histopathologic features of small melanomas. METHODS: The greatest diameters of 112 consecutively submitted MMs were measured histologically. Those lesions with diameters of less than 6 mm were further studied clinically and histopathologically. RESULTS: Four MMs (3.5%) were found to have small histologic diameters. Their clinical features differed from the classic clinical ABCD's, although their histopathologic features resembled those of large MMs. Their depth of dermal invasion was relatively superficial, and their clinical outcome was favorable. CONCLUSION: Histopathologically, small MMs tend to resemble large MMs, but clinically they may require other diagnostic criteria. Because they tend to show a relatively superficial depth of invasion, their increased recognition and removal may have an impact on overall mortality from MM.


Assuntos
Melanoma/patologia , Neoplasias Cutâneas/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias
15.
Mol Cell Biol ; 11(10): 5275-84, 1991 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1681425

RESUMO

Using Rous sarcoma virus as the vector, v-src or c-src genes were introduced into 6-day chicken embryo retina tissue in organ culture and their effects on retina development were investigated. Overexpression of c-src in many of the cells had no noticeable effect on retina development. In contrast, infection with v-src resulted in abnormal histogenesis and inhibition of differentiation. Although only a portion of the cells in infected tissue expressed the oncogene and displayed the transformation phenotype, the other cells were also hindered from becoming normally positioned and organized. Therefore, presence of oncogene-transformed cells within the tissue hindered organization and development of adjacent nontransformed cells. Failure of normal cell relationships impeded induction by cortisol of glutamine synthetase in Muller glia, which requires contact associations of the glia cells with neurons. The transformed cells tended to assemble into chaotic clusters, suggesting that their adhesiveness and contact affinities had become altered. This was confirmed by aggregation experiments with dissociated cells which showed that adhesiveness of transformed cells was greatly reduced and that they had lost the ability to cohere with nontransformed cells. In binary mixtures of transformed and nontransformed cells, the two sorted out into separate aggregates. Transformed cells formed loose clusters devoid of tissue architecture; aggregates of nontransformed cells became organized into retinotypic structures, and glutamine synthetase was inducible. Our findings suggest that the mechanisms of cell adhesion and cell affinities are a key target of v-src activity in infected cells and that modification of the cell surface may be a leading factor in other cellular changes characteristic of the v-src transformation phenotype.


Assuntos
Transformação Celular Neoplásica , Transformação Celular Viral , Genes src , Glutamato-Amônia Ligase/biossíntese , Retina/citologia , Animais , Vírus do Sarcoma Aviário/genética , Adesão Celular/genética , Diferenciação Celular/genética , Células Cultivadas , Embrião de Galinha , Indução Enzimática , Expressão Gênica , Glutamato-Amônia Ligase/genética , Técnicas de Cultura de Órgãos , Testes de Precipitina , Retina/embriologia , Retina/enzimologia , Retina/microbiologia
16.
J Virol ; 65(8): 4470-4, 1991 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-1649342

RESUMO

A cell-free system supporting transcription, replication, and nucleocapsid assembly of the genome RNA of human parainfluenza virus type 3 (HPF3) is described. Cytoplasmic extracts from infected CV-1 or BHK cells catalyzed the transcription of the entire HPF3 genome, the replication of genome RNA, and the assembly of this RNA into nucleocapsidlike structures. Newly replicated RNA was resistant to micrococcal nuclease digestion and was stable in CsCl gradients, exhibiting the density of authentic HPF3 nucleocapsids. After fractionation of the extracts, the nucleocapsid-containing pellet fraction synthesized viral mRNAs. Reconstitution with the soluble protein fraction was necessary for genome RNA replication and nucleocapsid assembly.


Assuntos
RNA Polimerases Dirigidas por DNA/metabolismo , Vírus da Parainfluenza 3 Humana/enzimologia , RNA Viral/biossíntese , RNA Polimerase Dependente de RNA/metabolismo , Transcrição Gênica , Animais , Capsídeo/biossíntese , Linhagem Celular , Centrifugação com Gradiente de Concentração , Fracionamento Químico , Eletroforese em Gel de Ágar , Humanos , Nuclease do Micrococo/metabolismo , Vírus da Parainfluenza 3 Humana/genética , Proteínas do Core Viral/biossíntese
17.
Virology ; 183(2): 821-4, 1991 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-1649512

RESUMO

CV-1 cell lines persistently infected with human parainfluenza virus type 3 (HPF3) contain one or more distinct subgenomic RNAs in addition to standard viral genomes. These RNAs are shown to be the genomes of defective-interfering (DI) particles of the virus; they are present in particles in the culture fluid, and they interfere with the growth of wild-type virus. Removal of the particles from the culture fluid by ultracentrifugation yields a supernatant fluid free from inhibitory activity, demonstrating that the anti-viral effect is not mediated by soluble factors. A role for the DI particles in persistence of HPF3 is considered.


Assuntos
Vírus Defeituosos/fisiologia , Vírus da Parainfluenza 3 Humana/fisiologia , Capsídeo/genética , Linhagem Celular , Vírus Defeituosos/genética , Vírus Defeituosos/metabolismo , Humanos , Vírus da Parainfluenza 3 Humana/genética , Vírus da Parainfluenza 3 Humana/metabolismo , RNA Viral/metabolismo , Solubilidade , Ultracentrifugação , Proteínas do Core Viral/genética , Replicação Viral/genética
18.
J Virol ; 65(6): 2773-7, 1991 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-1851852

RESUMO

Cells persistently infected with human parainfluenza virus type 3 (HPF3) exhibit a novel phenotype. They are completely resistant to fusion with each other but readily fuse with uninfected cells. We demonstrate that the inability of these cells to fuse with each other is due to a lack of cell surface neuraminic acid. Neuraminic acid is the receptor for the HPF3 hemagglutinin-neuraminidase (HN) glycoprotein, the molecule responsible for binding of the virus to cell surfaces. Uninfected CV-1 cells were treated with neuraminidase and then tested for their ability to fuse with the persistently infected (pi) cells. Neuraminidase treatment totally abolished cell fusion. To extend this result, we used a cell line deficient in sialic acid and demonstrated that these cells, like the neuraminidase-treated CV-1 cells, were unable to fuse with pi cells. We then tested whether mimicking the agglutinating function of the HN molecule with lectins would result in cell fusion. We added a panel of five lectins to the neuraminic acid-deficient cells and showed that binding of these cells to the pi cells did not result in fusion; the lectins could not substitute for interaction of neuraminic acid with the HN molecule in promoting membrane fusion. These results provide compelling evidence that the HN molecule of HPF3 and its interaction with neuraminic acid participate in membrane fusion and that cell fusion is mediated by an interaction more complex than mere juxtaposition of the cell membranes.


Assuntos
Proteína HN/farmacologia , Fusão de Membrana/efeitos dos fármacos , Vírus da Parainfluenza 3 Humana/fisiologia , Proteínas Virais de Fusão/farmacologia , Animais , Células Cultivadas/efeitos dos fármacos , Efeito Citopatogênico Viral , Humanos , Lectinas/farmacologia , Ácidos Neuramínicos/farmacologia , Infecções por Paramyxoviridae/patologia
19.
J Virol ; 64(7): 3212-8, 1990 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-2161938

RESUMO

Three cell lines persistently infected with human parainfluenza virus type 3 were characterized on a molecular level in this study. All six structural protein genes were transcribed into monocistronic RNAs in the persistently infected cells. In both acutely and persistently infected cells, polycistronic transcripts were abundant, although the ratio of polycistronic to monocistronic transcripts was reduced in the persistently infected cells. Each of the persistently infected cell lines contained a distinct subgenomic RNA species. The subgenomic RNAs were present in purified nucleocapsid cores, indicating that they represent viral genome RNA, were far more abundant than full-length RNA, and were stably maintained through at least 36 cell passages. Nucleotide sequence analysis of the subgenomic RNAs from two of the persistently infected cell lines revealed that the 5' ends are identical to that of the standard genome. Hybridization experiments with oligonucleotide probes showed that both fragments retain sequences from the 5' end of the standard genome and contain approximately 1,200 nucleotides (cell line 1) and 1,500 nucleotides (cell line 2) of the polymerase gene sequence. The demonstration of several alterations in viral gene expression in persistently infected cells offers insight into the factors associated with persistence of parainfluenza virus 3.


Assuntos
Vírus da Parainfluenza 3 Humana/genética , Infecções por Paramyxoviridae/microbiologia , Respirovirus/genética , Animais , Northern Blotting , Capsídeo/genética , Linhagem Celular , Células Cultivadas/microbiologia , Chlorocebus aethiops , Expressão Gênica , Genes Virais , Técnicas In Vitro , Peso Molecular , Sondas de Oligonucleotídeos , Vírus da Parainfluenza 3 Humana/crescimento & desenvolvimento , Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções por Paramyxoviridae/patologia , RNA Mensageiro/genética , RNA Viral/genética , Proteínas do Core Viral/genética , Proteínas Virais/biossíntese , Proteínas Virais/genética , Proteínas Estruturais Virais/genética , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA