Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 131(23)2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34609969

RESUMO

The capacity of respiratory viruses to undergo evolution within the respiratory tract raises the possibility of evolution under the selective pressure of the host environment or drug treatment. Long-term infections in immunocompromised hosts are potential drivers of viral evolution and development of infectious variants. We showed that intrahost evolution in chronic human parainfluenza virus 3 (HPIV3) infection in immunocompromised individuals elicited mutations that favored viral entry and persistence, suggesting that similar processes may operate across enveloped respiratory viruses. We profiled longitudinal HPIV3 infections from 2 immunocompromised individuals that persisted for 278 and 98 days. Mutations accrued in the HPIV3 attachment protein hemagglutinin-neuraminidase (HN), including the first in vivo mutation in HN's receptor binding site responsible for activating the viral fusion process. Fixation of this mutation was associated with exposure to a drug that cleaves host-cell sialic acid moieties. Longitudinal adaptation of HN was associated with features that promote viral entry and persistence in cells, including greater avidity for sialic acid and more active fusion activity in vitro, but not with antibody escape. Long-term infection thus led to mutations promoting viral persistence, suggesting that host-directed therapeutics may support the evolution of viruses that alter their biophysical characteristics to persist in the face of these agents in vivo.


Assuntos
Hospedeiro Imunocomprometido , Pneumopatias/virologia , Pulmão/virologia , Vírus da Parainfluenza 3 Humana/metabolismo , Infecções por Paramyxoviridae/virologia , Adulto , Sítios de Ligação , Análise Mutacional de DNA , Feminino , Frequência do Gene , Doença Enxerto-Hospedeiro/tratamento farmacológico , Células HEK293 , Humanos , Leucemia Mieloide Aguda , Mutação , Ácido Micofenólico/administração & dosagem , Ácido N-Acetilneuramínico/química , Vírus da Parainfluenza 3 Humana/genética , Infecções por Paramyxoviridae/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicações , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/virologia , Receptores Virais/metabolismo , Sirolimo/administração & dosagem , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus , Adulto Jovem
2.
J Virol ; 95(20): e0083721, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34319159

RESUMO

Interferon-induced transmembrane (IFITM) proteins inhibit a broad range of enveloped viruses by blocking entry into host cells. We used an inducible overexpression system to investigate if IFITM1, IFITM2, and IFITM3 could modulate early and/or late stages of influenza A virus (IAV) or parainfluenza virus 3 (PIV-3) infection in human A549 airway epithelial cells. IAV and PIV-3 represent respiratory viruses which utilize distinct cellular entry pathways. We verify entry by endocytosis for IAV, whereas PIV-3 infection was consistent with fusion at the plasma membrane. Following induction prior to infection, all three IFITM proteins restricted the percentage of IAV-infected cells at 8 hours postinfection. In contrast, prior induction of IFITM1 and IFITM2 did not inhibit PIV-3 infection, although a modest reduction was observed with IFITM3. Small interfering RNA (siRNA)-mediated knockdown of endogenous IFITM1, IFITM2, and IFITM3 expression, in the presence or absence of pretreatment with type I interferon, resulted in increased IAV, but not PIV-3, infection. This finding suggests that while all three IFITMs display antiviral activity against IAV, they do not restrict the early stages of PIV-3 infection. IAV and PIV-3 infection culminates in viral egress through budding at the plasma membrane. Inducible expression of IFITM1, IFITM2, or IFITM3 immediately after infection did not impact titers of infectious virus released from IAV- or PIV-3-infected cells. Our findings show that IFITM proteins differentially restrict the early stages of infection of two respiratory viruses with distinct cellular entry pathways but do not influence the late stages of replication for either virus. IMPORTANCE Interferon-induced transmembrane (IFITM) proteins restrict the initial stages of infection for several respiratory viruses; however, their potential to modulate the later stages of virus replication has not been explored. In this study, we highlight the utility of an inducible overexpression system to assess the impact of IFITM proteins on either early- or late-stage replication of two respiratory viruses. We demonstrate antiviral activity by IFITM1, IFITM2, and IFITM3 against influenza A virus (IAV) but not parainfluenza virus 3 (PIV-3) during the early stages of cellular infection. Furthermore, IFITM induction following IAV or PIV-3 infection does not restrict the late stages of replication of either virus. Our findings show that IFITM proteins can differentially restrict the early stages of infection of two viruses with distinct cellular entry pathways and yet do not influence the late stages of replication for either virus.


Assuntos
Viroses/metabolismo , Replicação Viral/fisiologia , Células A549 , Antígenos de Diferenciação/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Endocitose/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Vírus da Influenza A/metabolismo , Vírus da Influenza A/patogenicidade , Interferons/metabolismo , Proteínas de Membrana/metabolismo , Vírus da Parainfluenza 3 Humana/metabolismo , Vírus da Parainfluenza 3 Humana/patogenicidade , Proteínas de Ligação a RNA/metabolismo , Internalização do Vírus
3.
PLoS Pathog ; 16(9): e1008883, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32956394

RESUMO

Infection by human parainfluenza viruses (HPIVs) causes widespread lower respiratory diseases, including croup, bronchiolitis, and pneumonia, and there are no vaccines or effective treatments for these viruses. HPIV3 is a member of the Respirovirus species of the Paramyxoviridae family. These viruses are pleomorphic, enveloped viruses with genomes composed of single-stranded negative-sense RNA. During viral entry, the first step of infection, the viral fusion complex, comprised of the receptor-binding glycoprotein hemagglutinin-neuraminidase (HN) and the fusion glycoprotein (F), mediates fusion upon receptor binding. The HPIV3 transmembrane protein HN, like the receptor-binding proteins of other related viruses that enter host cells using membrane fusion, binds to a receptor molecule on the host cell plasma membrane, which triggers the F glycoprotein to undergo major conformational rearrangements, promoting viral entry. Subsequent fusion of the viral and host membranes allows delivery of the viral genetic material into the host cell. The intermediate states in viral entry are transient and thermodynamically unstable, making it impossible to understand these transitions using standard methods, yet understanding these transition states is important for expanding our knowledge of the viral entry process. In this study, we use cryo-electron tomography (cryo-ET) to dissect the stepwise process by which the receptor-binding protein triggers F-mediated fusion, when forming a complex with receptor-bearing membranes. Using an on-grid antibody capture method that facilitates examination of fresh, biologically active strains of virus directly from supernatant fluids and a series of biological tools that permit the capture of intermediate states in the fusion process, we visualize the series of events that occur when a pristine, authentic viral particle interacts with target receptors and proceeds from the viral entry steps of receptor engagement to membrane fusion.


Assuntos
Membrana Celular/metabolismo , Proteína HN/metabolismo , Vírus da Parainfluenza 3 Humana/metabolismo , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus , Animais , Membrana Celular/ultraestrutura , Chlorocebus aethiops , Humanos , Vírus da Parainfluenza 3 Humana/ultraestrutura , Células Vero
4.
Virus Genes ; 56(1): 37-48, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31768710

RESUMO

Human parainfluenza virus type 3 (HPIV3) causes the majority of childhood viral pneumonia around the world. Fusing the viral and target cell membranes is crucial for its entry into target cells, and the fusion process requires the concerted actions of two viral glycoproteins: hemagglutinin-neuraminidase (HN) and fusion (F) protein. After binding to the cell surface receptor, sialic acids, HN triggers F to undergo large conformational rearrangements to execute the fusion process. Although it has been reported that several domains of F had important impacts on regulating the membrane fusion activity, what role the DI-DII linker (residues 369-374, namely L1 linker) of the HPIV3 F protein plays in the fusion process still remains confused. We have obtained three chimeric mutant proteins (Ch-NDV-L1, Ch-MV-L1, Ch-HPIV1-L1) containing the full length of HPIV3 F protein but their corresponding DI-DII linker derived from the F protein of Newcastle disease virus (NDV), Measles virus (MV), and Human parainfluenza virus type 1 (HPIV1), respectively. One deletion mutant protein (De-L1), whose DI-DII linker was deleted, has been established simultaneously. Then vaccinia virus-T7 RNA polymerase transient expression system and standard plasmid system were utilized to express the mutant F proteins in BHK-21 cells. These four mutants were determined for membrane fusogenic activity, cell surface expression level, and total mutant F protein expression. All of them resulted in a significant reduction in fusogenic activity in all steps of cell-cell membrane fusion process. There was no significant difference in cell surface protein expression level for the mutants compared with wild-type F. The mutant proteins with inability in fusogenic activity were all at the form of precursor protein, F0, which were not hydrolyzed by intracellular protease furin. The results above suggest that the involvement of the DI-DII linker region is necessary for the complete fusion of the membranes.


Assuntos
Vírus da Parainfluenza 3 Humana/metabolismo , Infecções por Respirovirus/virologia , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus , Animais , Linhagem Celular , Membrana Celular/virologia , Proteína HN/genética , Proteína HN/metabolismo , Humanos , Fusão de Membrana , Vírus da Doença de Newcastle/genética , Vírus da Doença de Newcastle/metabolismo , Vírus da Parainfluenza 3 Humana/química , Vírus da Parainfluenza 3 Humana/genética , Domínios Proteicos , Proteínas Virais de Fusão/genética
5.
Biometals ; 31(1): 81-89, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29209895

RESUMO

Here we report on the results obtained from an antiviral screening, including herpes simplex virus, vaccinia virus, vesicular stomatitis virus, Coxsackie B4 virus or respiratory syncytial virus, parainfluenza-3 virus, reovirus-1 and Punta Toro virus, of three 2-hydroxy-3-methoxyphenyl acylhydrazone compounds in three cell lines (i.e. human embryonic lung fibroblast cells, human cervix carcinoma cells, and African Green monkey kidney cells). Interesting antiviral EC50 values are obtained against herpes simplex virus-1 and vaccinia virus. The biological activity of acylhydrazones is often attributed to their metal coordinating abilities, so potentiometric and microcalorimetric studies are here discussed to unravel the behavior of the three 2-hydroxy-3-methoxyphenyl compounds in solution. It is worth of note that the acylhydrazone with the higher affinity for Cu(II) ions shows the best antiviral activity against herpes simplex and vaccinia virus (EC50 ~ 1.5 µM, minimal cytotoxic concentration = 60 µM, selectivity index = 40).


Assuntos
Antivirais/farmacologia , Quelantes/farmacologia , Hidrazonas/farmacologia , Simplexvirus/efeitos dos fármacos , Vaccinia virus/efeitos dos fármacos , Animais , Antivirais/síntese química , Antivirais/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Quelantes/síntese química , Quelantes/metabolismo , Chlorocebus aethiops , Cobre/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/virologia , Humanos , Hidrazonas/síntese química , Hidrazonas/metabolismo , Concentração Inibidora 50 , Magnésio/metabolismo , Manganês/metabolismo , Orthoreovirus de Mamíferos/efeitos dos fármacos , Orthoreovirus de Mamíferos/crescimento & desenvolvimento , Orthoreovirus de Mamíferos/metabolismo , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Vírus da Parainfluenza 3 Humana/crescimento & desenvolvimento , Vírus da Parainfluenza 3 Humana/metabolismo , Phlebovirus/efeitos dos fármacos , Phlebovirus/crescimento & desenvolvimento , Phlebovirus/metabolismo , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Vírus Sinciciais Respiratórios/metabolismo , Simplexvirus/crescimento & desenvolvimento , Simplexvirus/metabolismo , Vaccinia virus/crescimento & desenvolvimento , Vaccinia virus/metabolismo , Células Vero , Vesiculovirus/efeitos dos fármacos , Vesiculovirus/crescimento & desenvolvimento , Vesiculovirus/metabolismo
6.
Virology ; 503: 46-51, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28113063

RESUMO

Interferon (IFN) exerts its antiviral effect by inducing a large family of cellular genes, named interferon (IFN)-stimulated genes (ISGs). An intriguing member of this family is indoleamine 2,3-dioxygenase (IDO), which catalyzes the first and rate-limiting step of the main branch of tryptophan (Trp) degradation, the kynurenine pathway. We recently showed that IDO strongly inhibits human parainfluenza virus type 3 (PIV3), a significant respiratory pathogen. Here, we show that 5-hydoxytryptophan (5-HTP), the first product of an alternative branch of Trp degradation and a serotonin precursor, is essential to protect virus growth against IDO in cell culture. We also show that the apparent antiviral effect of IDO on PIV3 is not due to the generation of the kynurenine pathway metabolites, but rather due to the depletion of intracellular Trp by IDO, as a result of which this rare amino acid becomes unavailable for the alternative, proviral 5-HTP pathway.


Assuntos
5-Hidroxitriptofano/metabolismo , Antivirais/farmacologia , Indolamina-Pirrol 2,3,-Dioxigenase/farmacologia , Vírus da Parainfluenza 3 Humana/crescimento & desenvolvimento , Triptofano/metabolismo , Replicação Viral/efeitos dos fármacos , 5-Hidroxitriptofano/farmacologia , Células A549 , Animais , Linhagem Celular Tumoral , Humanos , Interferons/farmacologia , Cinurenina/metabolismo , Macaca mulatta , Vírus da Parainfluenza 3 Humana/metabolismo , Infecções por Respirovirus/tratamento farmacológico , Triptofano/química , Replicação Viral/fisiologia
7.
Sci Rep ; 6: 24138, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-27053240

RESUMO

Human parainfluenza type-3 virus (hPIV-3) is one of the principal aetiological agents of acute respiratory illness in infants worldwide and also shows high disease severity in the elderly and immunocompromised, but neither therapies nor vaccines are available to treat or prevent infection, respectively. Using a multidisciplinary approach we report herein that the approved drug suramin acts as a non-competitive in vitro inhibitor of the hPIV-3 haemagglutinin-neuraminidase (HN). Furthermore, the drug inhibits viral replication in mammalian epithelial cells with an IC50 of 30 µM, when applied post-adsorption. Significantly, we show in cell-based drug-combination studies using virus infection blockade assays, that suramin acts synergistically with the anti-influenza virus drug zanamivir. Our data suggests that lower concentrations of both drugs can be used to yield high levels of inhibition. Finally, using NMR spectroscopy and in silico docking simulations we confirmed that suramin binds HN simultaneously with zanamivir. This binding event occurs most likely in the vicinity of the protein primary binding site, resulting in an enhancement of the inhibitory potential of the N-acetylneuraminic acid-based inhibitor. This study offers a potentially exciting avenue for the treatment of parainfluenza infection by a combinatorial repurposing approach of well-established approved drugs.


Assuntos
Células Epiteliais/efeitos dos fármacos , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Suramina/farmacologia , Zanamivir/farmacologia , Animais , Antivirais/metabolismo , Antivirais/farmacologia , Sítios de Ligação , Linhagem Celular , Sinergismo Farmacológico , Células Epiteliais/virologia , Proteína HN/metabolismo , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Rim/citologia , Cinética , Macaca mulatta , Espectroscopia de Ressonância Magnética , Simulação de Dinâmica Molecular , Vírus da Parainfluenza 3 Humana/metabolismo , Vírus da Parainfluenza 3 Humana/fisiologia , Ligação Proteica , Suramina/metabolismo , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos , Zanamivir/metabolismo
8.
PLoS One ; 11(2): e0148258, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26866481

RESUMO

Respiratory viruses are a cause of upper respiratory tract infections (URTI), but can be associated with severe lower respiratory tract infections (LRTI) in immunocompromised patients. The objective of this study was to investigate the genetic variability of influenza virus, parainfluenza virus and respiratory syncytial virus (RSV) and the duration of viral shedding in hematological patients. Nasopharyngeal swabs from hematological patients were screened for influenza, parainfluenza and RSV on admission as well as on development of respiratory symptoms. Consecutive swabs were collected until viral clearance. Out of 672 tested patients, a total of 111 patients (17%) were infected with one of the investigated viral agents: 40 with influenza, 13 with parainfluenza and 64 with RSV; six patients had influenza/RSV or parainfluenza/RSV co-infections. The majority of infected patients (n = 75/111) underwent stem cell transplantation (42 autologous, 48 allogeneic, 15 autologous and allogeneic). LRTI was observed in 48 patients, of whom 15 patients developed severe LRTI, and 13 patients with respiratory tract infection died. Phylogenetic analysis revealed a variety of influenza A(H1N1)pdm09, A(H3N2), influenza B, parainfluenza 3 and RSV A, B viruses. RSV A was detected in 54 patients, RSV B in ten patients. The newly emerging RSV A genotype ON1 predominated in the study cohort and was found in 48 (75%) of 64 RSV-infected patients. Furthermore, two distinct clusters were detected for RSV A genotype ON1, identical RSV G gene sequences in these patients are consistent with nosocomial transmission. Long-term viral shedding for more than 30 days was significantly associated with prior allogeneic transplantation (p = 0.01) and was most pronounced in patients with RSV infection (n = 16) with a median duration of viral shedding for 80 days (range 35-334 days). Long-term shedding of respiratory viruses might be a catalyzer of nosocomial transmission and must be considered for efficient infection control in immunocompromised patients.


Assuntos
Infecção Hospitalar/epidemiologia , Infecção Hospitalar/virologia , Doenças Hematológicas/virologia , Eliminação de Partículas Virais , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Coortes , Controle de Doenças Transmissíveis , Feminino , Genótipo , Humanos , Influenza Humana/virologia , Masculino , Pessoa de Meia-Idade , Mutação , Nariz/virologia , Orthomyxoviridae/metabolismo , Vírus da Parainfluenza 3 Humana/metabolismo , Infecções por Paramyxoviridae/virologia , Filogenia , Reação em Cadeia da Polimerase , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/metabolismo , Estudos Retrospectivos , Análise de Sequência de DNA , Fatores de Tempo , Transplante Homólogo , Adulto Jovem
9.
PLoS One ; 10(12): e0144648, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26659560

RESUMO

Fatty acid synthase (FASN) catalyzes the de novo synthesis of palmitate, a fatty acid utilized for synthesis of more complex fatty acids, plasma membrane structure, and post-translational palmitoylation of host and viral proteins. We have developed a potent inhibitor of FASN (TVB-3166) that reduces the production of respiratory syncytial virus (RSV) progeny in vitro from infected human lung epithelial cells (A549) and in vivo from mice challenged intranasally with RSV. Addition of TVB-3166 to the culture medium of RSV-infected A549 cells reduces viral spread without inducing cytopathic effects. The antiviral effect of the FASN inhibitor is a direct consequence of reducing de novo palmitate synthesis; similar doses are required for both antiviral activity and inhibition of palmitate production, and the addition of exogenous palmitate to TVB-3166-treated cells restores RSV production. TVB-3166 has minimal effect on RSV entry but significantly reduces viral RNA replication, protein levels, viral particle formation and infectivity of released viral particles. TVB-3166 substantially impacts viral replication, reducing production of infectious progeny 250-fold. In vivo, oral administration of TVB-3166 to RSV-A (Long)-infected BALB/c mice on normal chow, starting either on the day of infection or one day post-infection, reduces RSV lung titers 21-fold and 9-fold respectively. Further, TVB-3166 also inhibits the production of RSV B, human parainfluenza 3 (PIV3), and human rhinovirus 16 (HRV16) progeny from A549, HEp2 and HeLa cells respectively. Thus, inhibition of FASN and palmitate synthesis by TVB-3166 significantly reduces RSV progeny both in vitro and in vivo and has broad-spectrum activity against other respiratory viruses. FASN inhibition may alter the composition of regions of the host cell membrane where RSV assembly or replication occurs, or change the membrane composition of RSV progeny particles, decreasing their infectivity.


Assuntos
Antivirais/farmacologia , Inibidores Enzimáticos/farmacologia , Ácido Graxo Sintase Tipo I/antagonistas & inibidores , Processamento de Proteína Pós-Traducional , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Administração Oral , Animais , Antivirais/síntese química , Inibidores Enzimáticos/síntese química , Ácido Graxo Sintase Tipo I/genética , Ácido Graxo Sintase Tipo I/metabolismo , Expressão Gênica , Células HeLa , Células Hep G2 , Interações Hospedeiro-Patógeno , Humanos , Lipoilação/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Ácido Palmítico/antagonistas & inibidores , Ácido Palmítico/metabolismo , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Vírus da Parainfluenza 3 Humana/crescimento & desenvolvimento , Vírus da Parainfluenza 3 Humana/metabolismo , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/enzimologia , Mucosa Respiratória/virologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Vírus Sinciciais Respiratórios/metabolismo , Rhinovirus/efeitos dos fármacos , Rhinovirus/crescimento & desenvolvimento , Rhinovirus/metabolismo , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/genética , Proteínas Virais/metabolismo , Vírion/efeitos dos fármacos , Vírion/crescimento & desenvolvimento , Vírion/metabolismo
10.
mBio ; 4(5): e00803-13, 2013 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-24149514

RESUMO

UNLABELLED: Paramyxoviruses, enveloped RNA viruses that include human parainfluenza virus type 3 (HPIV3), cause the majority of childhood viral pneumonia. HPIV3 infection starts when the viral receptor-binding protein engages sialic acid receptors in the lung and the viral envelope fuses with the target cell membrane. Fusion/entry requires interaction between two viral surface glycoproteins: tetrameric hemagglutinin-neuraminidase (HN) and fusion protein (F). In this report, we define structural correlates of the HN features that permit infection in vivo. We have shown that viruses with an HN-F that promotes growth in cultured immortalized cells are impaired in differentiated human airway epithelial cell cultures (HAE) and in vivo and evolve in HAE into viable viruses with less fusogenic HN-F. In this report, we identify specific structural features of the HN dimer interface that modulate HN-F interaction and fusion triggering and directly impact infection. Crystal structures of HN, which promotes viral growth in vivo, show a diminished interface in the HN dimer compared to the reference strain's HN, consistent with biochemical and biological data indicating decreased dimerization and decreased interaction with F protein. The crystallographic data suggest a structural explanation for the HN's altered ability to activate F and reveal properties that are critical for infection in vivo. IMPORTANCE: Human parainfluenza viruses cause the majority of childhood cases of croup, bronchiolitis, and pneumonia worldwide. Enveloped viruses must fuse their membranes with the target cell membranes in order to initiate infection. Parainfluenza fusion proceeds via a multistep reaction orchestrated by the two glycoproteins that make up its fusion machine. In vivo, viruses adapt for survival by evolving to acquire a set of fusion machinery features that provide key clues about requirements for infection in human beings. Infection of the lung by parainfluenzavirus is determined by specific interactions between the receptor binding molecule (hemagglutinin-neuraminidase [HN]) and the fusion protein (F). Here we identify specific structural features of the HN dimer interface that modulate HN-F interaction and fusion and directly impact infection. The crystallographic and biochemical data point to a structural explanation for the HN's altered ability to activate F for fusion and reveal properties that are critical for infection by this important lung virus in vivo.


Assuntos
Proteína HN/metabolismo , Vírus da Parainfluenza 3 Humana/crescimento & desenvolvimento , Vírus da Parainfluenza 3 Humana/metabolismo , Infecções por Respirovirus/virologia , Proteínas Virais de Fusão/metabolismo , Proteínas Virais/metabolismo , Animais , Cristalografia por Raios X , Dimerização , Feminino , Proteína HN/química , Proteína HN/genética , Humanos , Vírus da Parainfluenza 3 Humana/enzimologia , Vírus da Parainfluenza 3 Humana/genética , Ligação Proteica , Ratos , Sigmodontinae , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética , Proteínas Virais/química , Proteínas Virais/genética
12.
J Biol Chem ; 287(1): 778-793, 2012 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-22110138

RESUMO

Parainfluenza viruses enter host cells by fusing the viral and target cell membranes via concerted action of their two envelope glycoproteins: the hemagglutinin-neuraminidase (HN) and the fusion protein (F). Receptor-bound HN triggers F to undergo conformational changes that render it fusion-competent. To address the role of receptor engagement and to elucidate how HN and F interact during the fusion process, we used bimolecular fluorescence complementation to follow the dynamics of human parainfluenza virus type 3 (HPIV3) HN/F pairs in living cells. We show that HN and F associate before receptor engagement. HN drives the formation of HN-F clusters at the site of fusion, and alterations in HN-F interaction determine the fusogenicity of the glycoprotein pair. An interactive site, at the HN dimer interface modulates HN fusion activation property, which is critical for infection of the natural host. This first evidence for the sequence of initial events that lead to viral entry may indicate a new paradigm for understanding Paramyxovirus infection.


Assuntos
Hemaglutininas Virais/metabolismo , Neuraminidase/metabolismo , Vírus da Parainfluenza 3 Humana/fisiologia , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus , Células HEK293 , Hemaglutininas Virais/química , Hemaglutininas Virais/genética , Humanos , Modelos Moleculares , Imagem Molecular , Mutação , Neuraminidase/química , Neuraminidase/genética , Vírus da Parainfluenza 3 Humana/metabolismo , Ligação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Receptores de Superfície Celular/metabolismo , Espectrometria de Fluorescência , Especificidade por Substrato
13.
Virus Genes ; 37(3): 358-67, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18751884

RESUMO

The P gene of human parainfluenza virus type 3 (HPIV 3) encodes a multicistronic P mRNA that gives rise to four polypeptides. The P and C proteins are synthesized from two discrete overlapping AUG codons from the unedited P mRNA, while synthesis of two additional proteins, V and PD, presumably occurs via a unique transcriptional editing mechanism. However, the presence of V and PD proteins in HPIV 3 infected cells and their role in viral replication remains uncertain. Here, in vitro expression of full-length PD protein from an altered P mRNA and generation of a polyclonal antibody to the COOH-terminus of PD was achieved. Confocal immunofluorescence analysis following Leptomycin B (LMB) treatment revealed the presence of PD protein in nuclear and cytoplasmic compartments of HPIV 3 infected cells suggesting the involvement of a nuclear localization signal in this process. These initial results provide new impetus for further characterization of the role of PD in HPIV 3 infection.


Assuntos
Expressão Gênica , Vírus da Parainfluenza 3 Humana/genética , Vírus da Parainfluenza 3 Humana/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoplasma/genética , Citoplasma/metabolismo , Células HeLa , Humanos , Fases de Leitura Aberta , Transporte Proteico
14.
Biochimie ; 89(6-7): 721-2, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17573176

RESUMO

I describe in this article the early days of interferon research in France. In 1957, when Isaacs and Lindenmann published their results in the same time we worked on a para-influenza 3 virus inhibitor. Thereafter we confirmed this important discovery and initiated research studies about anti-tumor action of interferon.


Assuntos
Interferons/química , Interferons/uso terapêutico , Vírus da Parainfluenza 3 Humana/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Antivirais/uso terapêutico , Biologia/história , História do Século XX , Humanos , Modelos Biológicos , Virologia/história
15.
Proc Natl Acad Sci U S A ; 104(23): 9585-90, 2007 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-17535916

RESUMO

RNase L, a principal mediator of innate immunity to viral infections in higher vertebrates, is required for a complete IFN antiviral response against certain RNA stranded viruses. dsRNA produced during viral infections activates IFN-inducible synthetases that produce 5'-phosphorylated, 2',5'-oligoadenylates (2-5A) from ATP. 2-5A activates RNase L in a wide range of different mammalian cell types, thus blocking viral replication. However, 2-5A has unfavorable pharmacologic properties; it is rapidly degraded, does not transit cell membranes, and leads to apoptosis. To obtain activators of RNase L with improved drug-like properties, high-throughput screening was performed on chemical libraries by using fluorescence resonance energy transfer. Seven compounds were obtained that activated RNase L at micromolar concentrations, and structure-activity relationship studies resulted in identification of an additional four active compounds. Two lead compounds were shown to have a similar mechanistic path toward RNase L activation as the natural activator 2-5A. The compounds bound to the 2-5A-binding domain of RNase L (as determined by surface plasmon resonance and confirmed by computational docking), and the compounds induced RNase L dimerization and activation. Interestingly, the low-molecular-weight activators of RNase L had broad-spectrum antiviral activity against diverse types of RNA viruses, including the human pathogen human parainfluenza virus type 3, yet these compounds by themselves were not cytotoxic at the effective concentrations. Therefore, these RNase L activators are prototypes for a previously uncharacterized class of broad-spectrum antiviral agents.


Assuntos
Antivirais/metabolismo , Endorribonucleases/metabolismo , Ativadores de Enzimas/metabolismo , Imunidade Inata/fisiologia , Vírus da Parainfluenza 3 Humana/metabolismo , Nucleotídeos de Adenina/metabolismo , Animais , Antivirais/farmacologia , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Dimerização , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Ativadores de Enzimas/farmacologia , Transferência Ressonante de Energia de Fluorescência , Camundongos , Modelos Moleculares , Oligonucleotídeos/genética , Oligorribonucleotídeos/metabolismo , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Ligação Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Ressonância de Plasmônio de Superfície , Replicação Viral/efeitos dos fármacos
16.
Vaccine ; 23(14): 1657-67, 2005 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-15705469

RESUMO

Human metapneumovirus (hMPV) infection causes respiratory tract disease similar to that observed during human respiratory syncytial virus infection (hRSV). hMPV infections have been reported across the entire age spectrum although the most severe disease occurs in young children. No vaccines, chemotherapeutics or antibodies are presently available for preventing or treating hMPV infections. In this study, a bovine/human chimeric parainfluenza virus type 3 (b/h PIV3) expressing the human parainfluenza type 3 (hPIV3) fusion (F) and hemagglutinin-neuraminidase (HN) proteins was engineered to express hMPV fusion (F) protein from the second genome position (b/h PIV3/hMPV F2) with the goal of generating a novel hMPV vaccine. b/h PIV3/hMPV F2 was previously shown to protect hamsters from challenge with wt hMPV (Tang RS, Schickli JH, Macphail M, Fernandes F, Bicha L, Spaete J, et al. Effects of human metapneumovirus and respiratory syncytial virus antigen insertion in two 3' proximal genome positions of bovine/human parainfluenza virus type 3 on virus replication and immunogenicity. J Virol 2003;77:10819-28) and is here further evaluated for efficacy and immunogenicity in African green monkeys (AGMs). AGMs immunized intranasally and intratracheally with b/h PIV3/hMPV F2 generated hMPV- and hPIV3-specific humoral and cellular immune responses and were protected from wt hMPV infection. In a separate study, the host-range restriction of b/h PIV3/hMPV F2 replication relative to wt hPIV3 was performed in rhesus monkeys to demonstrate attenuation. These studies showed that b/h PIV3/hMPV F2 was immunogenic, protective and attenuated in non-human primates and warrants further evaluation in humans as a vaccine candidate for prevention of hMPV-associated respiratory tract diseases.


Assuntos
Chlorocebus aethiops , Regulação Viral da Expressão Gênica/imunologia , Metapneumovirus/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Paramyxoviridae/prevenção & controle , Proteínas Virais de Fusão/biossíntese , Proteínas Virais de Fusão/imunologia , Animais , Humanos , Macaca mulatta , Metapneumovirus/metabolismo , Vírus da Parainfluenza 3 Humana/metabolismo , Infecções por Paramyxoviridae/imunologia , Infecções por Paramyxoviridae/virologia , Células Vero , Proteínas Virais de Fusão/genética
17.
Biotechnol Prog ; 20(6): 1810-6, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15575716

RESUMO

We describe the generation of lentiviruses pseudotyped with human parainfluenza type 3 envelope (HPIV3) glycoproteins. Lentivirus particles, expressed in 293T/17 cells, incorporate HPIV3 hemagglutinin-neuraminidase (HN) and fusion (F) proteins into their lipid bilayers and are able to transduce human kidney epithelial cells and polarized MDCK cells. Neuraminidase, AZT, and anti-HPIV3 antisera block transduction, which is consistent with lentiviral-mediated transduction via sialated receptors for HPIV3. Our findings show that HPIV3 pseudotyped lentiviruses can be formed and may have a number of useful properties for human gene transfer.


Assuntos
Vetores Genéticos/genética , Proteína HN/metabolismo , Rim/metabolismo , Lentivirus/genética , Vírus da Parainfluenza 3 Humana/metabolismo , Transdução Genética/métodos , Proteínas Virais de Fusão/metabolismo , Linhagem Celular , Proteína HN/genética , Humanos , Vírus da Parainfluenza 3 Humana/genética , Engenharia de Proteínas/métodos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Virais de Fusão/genética
18.
Immunol Res ; 30(2): 125-38, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15477655

RESUMO

The major histocompatibility complex (MHC) class II transactivator (CIITA) typically is required for both constitutive and inducible expression of MHC class II genes. However, transcription of class II MHC genes has been observed in specific cell types (e.g., thymic epithelial cells) in CIITA-deficient mice as well as in specific situations (e.g., following viral infections or in natural killer [NK]/target cell interaction). These observations have been interpreted by some to indicate that a CIITA-independent pathway of class II gene expression might be germane to processes such as the acquisition of tolerance during thymic selection or in the evasion of immune surveillance by a subset of viruses. One of the most striking examples of CIITA-independent, inducible class II gene expression has involved the de novo expression of class II MHC molecules on respiratory epithelial cells following infection by human parainfluenza virus type 3 (HPIV3). We report here that despite careful analysis using multiple techniques, we have been unable to detect HPIV3-dependent, CIITA-independent (or CIITA-dependent) induction of class II MHC genes. Thus, whereas there may still be an intriguing role for CIITA-independent gene expression in facets of the immune response, this is unlikely to manifest in the analysis of HPIV3 infection of respiratory epithelial cells.


Assuntos
Células Epiteliais/metabolismo , Antígenos de Histocompatibilidade Classe II/biossíntese , Proteínas Nucleares/metabolismo , Vírus da Parainfluenza 3 Humana/metabolismo , Sistema Respiratório/metabolismo , Infecções por Respirovirus/metabolismo , Transativadores/metabolismo , Animais , Linhagem Celular , Células Epiteliais/virologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Antígenos de Histocompatibilidade Classe II/genética , Humanos , Camundongos , Vírus da Parainfluenza 3 Humana/genética , Sistema Respiratório/citologia , Sistema Respiratório/virologia , Infecções por Respirovirus/patologia , Infecções por Respirovirus/virologia
19.
Virology ; 298(1): 73-83, 2002 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-12093175

RESUMO

Our current studies have demonstrated that human parainfluenza virus type 3 (HPIV-3) utilizes heparan sulfate (HS) for its efficient cellular entry. HPIV-3 interacted with HS-agarose in vitro and the cellular entry and infection of HPIV-3 were reduced following (a) infection of human epithelial lung A549 cells with HPIV-3 pre-incubated with soluble HS; (b) treatment of A549 cells with heparinase to remove cell surface HS and sodium chlorate (NaClO(3)), a potent inhibitor of proteoglycan sulfation; and (c) infection of HS-deficient mutant CHO cell lines. However, in each instance, complete inhibition of HPIV-3 entry did not occur, suggesting the presence of additional nonproteoglycan cell surface molecule(s) that is required for HPIV-3 entry. Thus the cell surface HS appears to play an important role in efficient cellular entry of HPIV-3.


Assuntos
Células Epiteliais/virologia , Heparitina Sulfato/fisiologia , Vírus da Parainfluenza 3 Humana/fisiologia , Animais , Células CHO/virologia , Membrana Celular/virologia , Cricetinae , Heparina Liase/farmacologia , Heparitina Sulfato/farmacologia , Humanos , Pulmão , Vírus da Parainfluenza 3 Humana/metabolismo , Replicação Viral
20.
J Virol ; 75(10): 4604-13, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11312330

RESUMO

Through their hemagglutinin-neuraminidase glycoprotein, parainfluenza viruses bind to sialic acid-containing glycoconjugates to initiate infection. Although the virus-receptor interaction is a key factor of infection, the exact nature of the receptors that human parainfluenza viruses recognize has not been determined. We evaluated the abilities of human parainfluenza virus types 1 (hPIV-1) and 3 (hPIV-3) to bind to different types of gangliosides. Both hPIV-1 and hPIV-3 preferentially bound to neolacto-series gangliosides containing a terminal N-acetylneuraminic acid (NeuAc) linked to N-acetyllactosamine (Galbeta1-4GlcNAc) by the alpha2-3 linkage (NeuAcalpha2-3Galbeta1-4GlcNAc). Unlike hPIV-1, hPIV-3 bound to gangliosides with a terminal NeuAc linked to Galbeta1-4GlcNAc through an alpha2-6 linkage (NeuAcalpha2-6Galbeta1-4GlcNAc) or to gangliosides with a different sialic acid, N-glycolylneuraminic acid (NeuGc), linked to Galbeta1-4GlcNAc (NeuGcalpha2-3Galbeta1-4GlcNAc). These results indicate that the molecular species of glycoconjugate that hPIV-1 recognizes are more limited than those recognized by hPIV-3. Further analysis using purified gangliosides revealed that the oligosaccharide core structure is also an important element for binding. Gangliosides that contain branched N-acetyllactosaminoglycans in their core structure showed higher avidity than those without them. Agglutination of human, cow, and guinea pig erythrocytes but not equine erythrocytes by hPIV-1 and hPIV-3 correlated well with the presence or the absence of sialic acid-linked branched N-acetyllactosaminoglycans on the cell surface. Finally, NeuAcalpha2-3I, which bound to both viruses, inhibited virus infection of Lewis lung carcinoma-monkey kidney cells in a dose-dependent manner. We conclude that hPIV-1 and hPIV-3 preferentially recognize oligosaccharides containing branched N-acetyllactosaminoglycans with terminal NeuAcalpha2-3Gal as receptors and that hPIV-3 also recognizes NeuAcalpha2-6Gal- or NeuGcalpha2-3Gal-containing receptors. These findings provide important information that can be used to develop inhibitors that prevent human parainfluenza virus infection.


Assuntos
Gangliosídeos/metabolismo , Vírus da Parainfluenza 1 Humana/metabolismo , Vírus da Parainfluenza 3 Humana/metabolismo , Receptores Virais/metabolismo , Animais , Sequência de Carboidratos , Galactose/metabolismo , Humanos , Macaca mulatta , Dados de Sequência Molecular , Ácido N-Acetilneuramínico/metabolismo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA