Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 383
Filtrar
1.
PLoS Pathog ; 17(9): e1009908, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34529742

RESUMO

Human parainfluenza virus type 1 (hPIV1) and 3 (hPIV3) cause seasonal epidemics, but little is known about their interaction with human airway cells. In this study, we determined cytopathology, replication, and progeny virion release from human airway cells during long-term infection in vitro. Both viruses readily established persistent infection without causing significant cytopathic effects. However, assembly and release of hPIV1 rapidly declined in sharp contrast to hPIV3 due to impaired viral ribonucleocapsid (vRNP) trafficking and virus assembly. Transcriptomic analysis revealed that both viruses induced similar levels of type I and III IFNs. However, hPIV1 induced specific ISGs stronger than hPIV3, such as MX2, which bound to hPIV1 vRNPs in infected cells. In addition, hPIV1 but not hPIV3 suppressed genes involved in lipid biogenesis and hPIV1 infection resulted in ubiquitination and degradation of 3-hydroxy-3-methylglutaryl-coenzyme A reductase, a rate limiting enzyme in cholesterol biosynthesis. Consequently, formation of cholesterol-rich lipid rafts was impaired in hPIV1 infected cells. These results indicate that hPIV1 is capable of regulating cholesterol biogenesis, which likely together with ISGs contributes to establishment of a quiescent infection.


Assuntos
Colesterol/biossíntese , Mucosa Respiratória/virologia , Infecções por Respirovirus/metabolismo , Infecções por Respirovirus/virologia , Células A549 , Humanos , Interferons/imunologia , Vírus da Parainfluenza 1 Humana/imunologia , Vírus da Parainfluenza 1 Humana/metabolismo , Vírus da Parainfluenza 3 Humana/imunologia , Vírus da Parainfluenza 3 Humana/metabolismo , Infecções por Respirovirus/imunologia
2.
Pol J Vet Sci ; 24(2): 235-241, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34250778

RESUMO

Respiratory diseases constitute a major health problem in small ruminant herds around the world, and parainfluenza virus type 3 (PIV-3) has been shown to play a vital role in their etiology. This cross-sectional study describes the serological status of the non-vaccinated dairy goat popu- lation in Poland with respect to PIV-3 infection and investigates the relationship between the presence of antibodies to PIV-3 and some basic herd-level and animal-level factors, including small ruminant lentivirus (SRLV) infection. Serum samples from 1188 goats from 48 herds were tested for the concentration of antibodies to PIV-3 using a quantitative immunoenzymatic assay. Specific antibodies were detected in all tested goats from all herds. The concentration of PIV-3 antibodies varied from 8.4 to >240 ng/ml (median 95.9 ng/ml) and was significantly higher in goats from larger herds and from these herds in which cough was often observed by farmers. Moreover, it was noted that female goats had higher antibody concentrations than males. On the other hand, the concentration of PIV-3 antibodies did not prove to be significantly linked to the presence of SRLV infection. This study shows that PIV-3 infection in the Polish goat population is widespread and appears to contribute to the occurrence of respiratory diseases in goat herds.


Assuntos
Anticorpos Antivirais/sangue , Doenças das Cabras/virologia , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Respirovirus/veterinária , Animais , Estudos Transversais , Feminino , Doenças das Cabras/epidemiologia , Cabras , Masculino , Polônia/epidemiologia , Infecções por Respirovirus/epidemiologia , Infecções por Respirovirus/virologia
3.
MAbs ; 13(1): 1912884, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33876699

RESUMO

Human parainfluenza virus type III (HPIV3) is a common respiratory pathogen that afflicts children and can be fatal in vulnerable populations, including the immunocompromised. There are currently no effective vaccines or therapeutics available, resulting in tens of thousands of hospitalizations per year. In an effort to discover a protective antibody against HPIV3, we screened the B cell repertoires from peripheral blood, tonsils, and spleen from healthy children and adults. These analyses yielded five monoclonal antibodies that potently neutralized HPIV3 in vitro. These HPIV3-neutralizing antibodies targeted two non-overlapping epitopes of the HPIV3 F protein, with most targeting the apex. Prophylactic administration of one of these antibodies, PI3-E12, resulted in potent protection against HPIV3 infection in cotton rats. Additionally, PI3-E12 could also be used therapeutically to suppress HPIV3 in immunocompromised animals. These results demonstrate the potential clinical utility of PI3-E12 for the prevention or treatment of HPIV3 in both immunocompetent and immunocompromised individuals.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Antivirais/farmacologia , Pulmão/virologia , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Infecções por Respirovirus/prevenção & controle , Proteínas Virais de Fusão/antagonistas & inibidores , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Especificidade de Anticorpos , Antivirais/imunologia , Linfócitos B/imunologia , Linfócitos B/virologia , Linhagem Celular , Modelos Animais de Doenças , Epitopos , Interações Hospedeiro-Patógeno , Humanos , Hospedeiro Imunocomprometido , Pulmão/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/virologia , Sigmodontinae , Proteínas Virais de Fusão/imunologia
4.
Vet Microbiol ; 254: 108980, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33445054

RESUMO

The Caprine parainfluenza virus 3 (CPIV3) is a novel Paramyxovirus that is isolated from goats suffering from respiratory diseases. Presently, the pathogenesis of CPIV3 infection has not yet been fully characterized. The Type I interferon (IFN) is a key mediator of innate antiviral responses, as many viruses have developed strategies to circumvent IFN response, whether or how CPIV3 antagonizes type I IFN antiviral effects have not yet been characterized. This study observed that CPIV3 was resistant to IFN-α treatment and antagonized IFN-α antiviral responses on MDBK and goat tracheal epithelial (GTE) cell models. Western blot analysis showed that CPIV3 infection reduced STAT1 expression and phosphorylation, which inhibited IFN-α signal transduction on GTE cells. By screening and utilizing specific monoclonal antibodies (mAbs), three CPIV3 accessory proteins C, V and D were identified during the virus infection process on the GTE cell models. Accessory proteins C and V, but not protein D, was identified to antagonize IFN-α antiviral signaling. Furthermore, accessory protein C, but not protein V, reduced the level of IFN-α driven phosphorylated STAT1 (pSTAT1), and then inhibit STAT1 signaling. Genetic variation analysis to the PIV3 accessory protein C has found two highly variable regions (VR), with VR2 (31-70th aa) being involved in for the CPIV3 accessory protein C to hijack the STAT1 signaling activation. The above data indicated that CPIV3 is capable of inhibiting IFN-α signal transduction by reducing STAT1 expression and activation, and that the accessory protein C, plays vital roles in the immune escape process.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Antivirais/farmacologia , Evasão da Resposta Imune , Interferon Tipo I/antagonistas & inibidores , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Vírus da Parainfluenza 3 Humana/genética , Infecções por Paramyxoviridae/veterinária , Animais , Anticorpos Monoclonais/farmacologia , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Feminino , Doenças das Cabras/virologia , Cabras/virologia , Células HEK293 , Ensaios de Triagem em Larga Escala , Humanos , Interferon-alfa/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Paramyxoviridae/tratamento farmacológico , Fosforilação , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT1/genética , Transdução de Sinais/efeitos dos fármacos
5.
Front Immunol ; 11: 575977, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33123159

RESUMO

Human Parainfluenza Virus-3 (HPIV3) causes severe respiratory illness in immunocompromised patients and lacks approved anti-viral therapies. A phase I study of adoptively transferred virus-specific T-cells (VSTs) targeting HPIV3 following bone marrow transplantation is underway (NCT03180216). We sought to identify immunodominant epitopes within HPIV3 Matrix protein and their cross-reactivity against related viral proteins. VSTs were generated from peripheral blood of healthy donors by ex-vivo expansion after stimulation with a 15-mer peptide library encompassing HPIV3 matrix protein. Epitope mapping was performed using IFN-γ ELIspot with combinatorial peptide pools. Flow cytometry was used to characterize products with intracellular cytokine staining. In 10 VST products tested, we discovered 12 novel immunodominant epitopes. All products recognized an epitope at the C-terminus. On IFN-γ ELISpot, individual peptides eliciting activity demonstrated mean IFN-γ spot forming units per well (SFU)/1x105 cells of 115.5 (range 24.5-247.5). VST products were polyfunctional, releasing IFN-γ and TNF-α in response to identified epitopes, which were primarily HLA Class II restricted. Peptides from Human Parainfluenza Virus-1 corresponding to the HPIV3 epitopes showed cross-reactivity for HPIV1 in 11 of 12 tested epitopes (mean cross reactivity index: 1.19). Characterization of HPIV3 epitopes may enable development of third-party VSTs to treat immune suppressed patients with HPIV infection.


Assuntos
Transferência Adotiva , Epitopos Imunodominantes , Vírus da Parainfluenza 1 Humana/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Respirovirus/terapia , Linfócitos T/transplante , Proteínas da Matriz Viral/imunologia , Células Cultivadas , Ensaios Clínicos Fase I como Assunto , Reações Cruzadas , ELISPOT , Mapeamento de Epitopos , Interações Hospedeiro-Patógeno , Humanos , Interferon gama/metabolismo , Testes de Liberação de Interferon-gama , Vírus da Parainfluenza 1 Humana/patogenicidade , Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/metabolismo , Infecções por Respirovirus/virologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
6.
BMC Infect Dis ; 20(1): 353, 2020 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-32429853

RESUMO

BACKGROUND: The aim of the study was to identify the pathogens, in addition to bordetella pertussis (B. pertussis), which cause pertussis-like syndrome in children and to compare clinical presentation between those with B. pertussis and pertussis-like syndrome. METHODS: A cross-sectional analysis was conducted from March 2016 to September 2018. In total, 281 children with suspected pertussis infections were enrolled in this study. Multi-pathogen detection was performed. RESULTS: In total, 281 children were enrolled including 139 males and 142 females. Among them, 149 (53.0%) were B. pertussis positive, and 72 (15.6%) children tested positive for other pathogens. Mycoplasma pneumoniae (MP, 27 cases) was the most common causative pathogen in pertussis-like syndrome, followed by human rhinovirus (HRV, 23 cases), Streptococcus pneumoniae (SP, 13 cases), Haemophilus influenzae (HI, 12 cases) and parainfluenza virus 3 (Pinf-3, 9 cases). Children in the B. pertussis group had a higher rate of vaccination and longer hospital stay (P < 0.05). B. pertussis was more likely to be detected in winter than other pathogens, but this difference was not significant (P = 0.074). The number of white blood cells, neutrophils and blood platelets was significantly higher in children in the B. pertussis than in the pertussis-like group (P < 0.05). In addition, the percentage of CD3-CD19+ cells was significantly higher in the B. pertussis group (P = 0.018). CONCLUSION: About half of the children with pertussis-like syndrome were B. pertussis positive. MP was the second most common causative pathogen followed by HRV, SP, HI and Pinf-3. Children infected with B. pertussis had longer hospital stay and higher numbers of white blood cells, neutrophil and blood platelets compared with other pathogens.


Assuntos
Bordetella pertussis/genética , Haemophilus influenzae/genética , Mycoplasma pneumoniae/genética , Vírus da Parainfluenza 3 Humana/imunologia , Rhinovirus/genética , Streptococcus pneumoniae/genética , Coqueluche/diagnóstico , Criança , Pré-Escolar , Estudos Transversais , Feminino , Imunofluorescência , Humanos , Lactente , Recém-Nascido , Tempo de Internação , Contagem de Leucócitos , Masculino , Neutrófilos , Contagem de Plaquetas , Reação em Cadeia da Polimerase em Tempo Real , Síndrome , Coqueluche/virologia
7.
J Immunol ; 205(1): 261-271, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32423918

RESUMO

IFNs, produced during viral infections, induce the expression of hundreds of IFN-stimulated genes (ISGs). Some ISGs have specific antiviral activity, whereas others regulate the cellular response. Besides functioning as an antiviral effector, ISG15 is a negative regulator of IFN signaling, and inherited ISG15 deficiency leads to autoinflammatory IFNopathies, in which individuals exhibit elevated ISG expression in the absence of pathogenic infection. We have recapitulated these effects in cultured human A549-ISG15-/- cells and (using A549-UBA7-/- cells) confirmed that posttranslational modification by ISG15 (ISGylation) is not required for regulation of the type I IFN response. ISG15-deficient cells pretreated with IFN-α were resistant to paramyxovirus infection. We also showed that IFN-α treatment of ISG15-deficient cells led to significant inhibition of global protein synthesis, leading us to ask whether resistance was due to the direct antiviral activity of ISGs or whether cells were nonpermissive because of translation defects. We took advantage of the knowledge that IFN-induced protein with tetratricopeptide repeats 1 (IFIT1) is the principal antiviral ISG for parainfluenza virus 5. Knockdown of IFIT1 restored parainfluenza virus 5 infection in IFN-α-pretreated, ISG15-deficient cells, confirming that resistance was due to the direct antiviral activity of the IFN response. However, resistance could be induced if cells were pretreated with IFN-α for longer times, presumably because of inhibition of protein synthesis. These data show that the cause of virus resistance is 2-fold; ISG15 deficiency leads to the early overexpression of specific antiviral ISGs, but the later response is dominated by an unanticipated, ISG15-dependent loss of translational control.


Assuntos
Citocinas/deficiência , Resistência à Doença/genética , Interferon-alfa/metabolismo , Infecções por Paramyxoviridae/imunologia , Transdução de Sinais/imunologia , Ubiquitinas/deficiência , Células A549 , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Chlorocebus aethiops , Técnicas de Silenciamento de Genes , Técnicas de Inativação de Genes , Humanos , Vírus da Parainfluenza 2 Humana/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Vírus da Parainfluenza 5/imunologia , Infecções por Paramyxoviridae/virologia , Processamento de Proteína Pós-Traducional/imunologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais/genética , Enzimas Ativadoras de Ubiquitina/genética , Células Vero
8.
PLoS One ; 15(2): e0228572, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32045432

RESUMO

Human respiratory syncytial virus (RSV) and parainfluenza virus type 3 (HPIV3) are among the most common viral causes of childhood bronchiolitis and pneumonia worldwide, and lack effective antiviral drugs or vaccines. Recombinant (r) HPIV3 was modified to express the RSV fusion (F) glycoprotein, the major RSV neutralization and protective antigen, providing a live intranasal bivalent HPIV3/RSV vaccine candidate. This extends previous studies using a chimeric bovine-human PIV3 vector (rB/HPIV3). One advantage is that rHPIV3 expresses all of the HPIV3 antigens compared to only two for rB/HPIV3. In addition, the use of rHPIV3 as vector should avoid excessive attenuation following addition of the modified RSV F gene, which may occur with rB/HPIV3. To enhance its immunogenicity, RSV F was modified (i) to increase the stability of the prefusion (pre-F) conformation and (ii) by replacement of its transmembrane (TM) and cytoplasmic tail (CT) domains with those of HPIV3 F (H3TMCT) to increase incorporation in the vector virion. RSV F (+/- H3TMCT) was expressed from the first (F/preN) or the second (F/N-P) gene position of rHPIV3. The H3TMCT modification dramatically increased packaging of RSV F into the vector virion and, in hamsters, resulted in significant increases in the titer of high-quality serum RSV-neutralizing antibodies, in addition to the increase conferred by pre-F stabilization. Only F-H3TMCT/preN replication was significantly attenuated in the nasal turbinates by the RSV F insert. F-H3TMCT/preN, F/N-P, and F-H3TMCT/N-P provided complete protection against wt RSV challenge. F-H3TMCT/N-P exhibited the most stable and highest expression of RSV F, providing impetus for its further development.


Assuntos
Vacinas contra Parainfluenza/genética , Vírus da Parainfluenza 3 Humana/imunologia , Vacinas contra Vírus Sincicial Respiratório/genética , Proteínas Virais de Fusão/genética , Montagem de Vírus , Administração Intranasal , Animais , Chlorocebus aethiops , Cricetinae , Feminino , Humanos , Imunogenicidade da Vacina , Macaca mulatta , Mesocricetus , Vacinas contra Parainfluenza/administração & dosagem , Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 3 Humana/genética , Vírus da Parainfluenza 3 Humana/fisiologia , Estabilidade Proteica , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/imunologia , Células Vero , Proteínas Virais de Fusão/metabolismo
9.
Vet Microbiol ; 241: 108573, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31928705

RESUMO

Caprine parainfluenza virus type 3 (CPIV3) is the one of most common causative agents of caprine respiratory infection, resulting in significant economic losses in the goat and sheep industries. However, the molecular mechanisms and host genes involved in the pathogenesis of and immunity against CPIV3 infection remain poorly understood. In this study, we used RNA-Seq to understand the responses of madin-darby bovine kidney (MDBK) cells to CPIV3 infection. A total of 261 differentially-expressed genes (DEGs) were identified in CPIV3-infected compared with mock-infected MDBK cells at 24 h post-infection (hpi). The DEGs were mainly involved in immune system processes, metabolic processes, and signal transduction. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis demonstrated that the most significantly enriched signaling pathways were MAPK, Wnt, PI3K-Akt, tumor necrosis factor, Toll-like receptor and ubiquitin-mediated proteolysis. STRING analysis revealed that seven interferon-stimulated genes (ISGs) were upregulated (IFI6, ISG15, OAS1Y, OAS1Z, MX1, MX2 and RSAD2) and may play a pivotal role during CPIV3 infection. Moreover, overexpression of these ISGs significantly reduced CPIV3 replication in vitro, while siRNA silencing markedly improved CPIV3 replication 24 and 48 hpi. Ours is the first study to profile the gene expression of CPIV3-infected MDBK cells. We identified seven ISGs that could be targeted in novel antiviral strategies against CPIV3.


Assuntos
Interferons/farmacologia , Vírus da Parainfluenza 3 Humana/fisiologia , Replicação Viral , Animais , Bovinos , Linhagem Celular , Cães , Técnica Indireta de Fluorescência para Anticorpo/veterinária , Regulação Viral da Expressão Gênica , Técnicas de Silenciamento de Genes/veterinária , Cabras , Microesferas , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Vírus da Parainfluenza 3 Humana/genética , Vírus da Parainfluenza 3 Humana/imunologia , RNA Viral/química , RNA Viral/isolamento & purificação , Ensaio de Radioimunoprecipitação/veterinária , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Transcriptoma , Replicação Viral/efeitos dos fármacos , Replicação Viral/imunologia
10.
Biomed Res Int ; 2020: 2616172, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33457403

RESUMO

Human parainfluenza virus type 3 (HPIV3) is the main pathogen that causes respiratory infections in infants, young children, and the elderly. Currently, there are no vaccines and effective anti-infective drugs. Studying the replication and proliferation mechanism of HPIV3 is helpful for exploring the targets of anti-HPIV3 infection. Matrix protein (M) and nucleocapsid protein (N) are two key structural proteins of HPIV3 that exert important functions in HPIV3 proliferation. Herein, we aim to clarify the functional domains of M and N interaction. HPIV3 M and N expression plasmids of pCAGGS-HA-M and pCAGGS-N-Myc/Flag, M C-terminal truncation mutant plasmids of pCAGGSHA-MΔC120, MΔC170, MΔC190, and MΔC210, and M C-terminal plasmid of pCAGGS-HA-MC190 and C-terminal deletion mutant plasmid of pCAGGS-MΔN143-182 were constructed. By using immunoprecipitation, immunofluorescence, and virus-like particle (VLP) germination experiments, we found that N was encapsulated into M-mediated VLP through N and M interaction. Moreover, the C-terminus of the M played a key role in the interaction between M and N. The C-terminus of the M encapsulated the N into the VLP. We finally determined that the 143-182 amino acids in the M were the functional regions that encapsulated the N into the M-mediated VLP. Our findings confirmed the interaction between M and N and for the first time clarified that the 143-182 amino acid region in M was the functional region that interacted with N, which provides a molecular basis for exploring effective anti-HPIV3 targets.


Assuntos
Proteínas do Nucleocapsídeo/metabolismo , Vírus da Parainfluenza 3 Humana/imunologia , Vírus da Parainfluenza 3 Humana/metabolismo , Proteínas da Matriz Viral/metabolismo , Replicação Viral , Membrana Celular/metabolismo , Citoplasma/metabolismo , Deleção de Genes , Células HEK293 , Células HeLa , Humanos , Microscopia de Fluorescência , Plasmídeos/metabolismo , Ligação Proteica , Domínios Proteicos
11.
Cells ; 8(10)2019 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-31575075

RESUMO

DEAD-box helicase 3, X-linked (DDX3X) regulates the retinoic acid-inducible gene I (RIG-I)-like receptor (RLR)-mediated antiviral response, but can also be a host factor contributing to the replication of viruses of significance to human health, such as human immunodeficiency virus type 1 (HIV-1). These roles are mediated in part through its ability to actively shuttle between the nucleus and the cytoplasm to modulate gene expression, although the trafficking mechanisms, and impact thereof on immune signaling and viral infection, are incompletely defined. We confirm that DDX3X nuclear export is mediated by the nuclear transporter exportin-1/CRM1, dependent on an N-terminal, leucine-rich nuclear export signal (NES) and the monomeric guanine nucleotide binding protein Ran in activated GTP-bound form. Transcriptome profiling and ELISA show that exportin-1-dependent export of DDX3X to the cytoplasm strongly impacts IFN-ß production and the upregulation of immune genes in response to infection. That this is key to DDX3X's antiviral role was indicated by enhanced infection by human parainfluenza virus-3 (hPIV-3)/elevated virus production when the DDX3X NES was inactivated. Our results highlight a link between nucleocytoplasmic distribution of DDX3X and its role in antiviral immunity, with strong relevance to hPIV-3, as well as other viruses such as HIV-1.


Assuntos
Núcleo Celular/metabolismo , RNA Helicases DEAD-box/metabolismo , Carioferinas/fisiologia , Vírus da Parainfluenza 3 Humana/imunologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Infecções por Respirovirus/imunologia , Células A549 , Transporte Ativo do Núcleo Celular , Animais , Chlorocebus aethiops , Citoplasma/metabolismo , Células HEK293 , Células HeLa , Humanos , Carioferinas/genética , Receptores Citoplasmáticos e Nucleares/genética , Células Vero , Proteína Exportina 1
12.
Artigo em Alemão | MEDLINE | ID: mdl-31212341

RESUMO

OBJECTIVE: Bovine respiratory diseases are a common cause of calf loss. This study aimed to analyse associations between an occurrence of enzootic bronchopneumonia (EBP), calf mortality and calving management. MATERIAL AND METHODS: A total of 153 dairy farms participated in the study on a voluntary basis from November 2006 to July 2007. Calf management was inspected on-site during a farm visit and farm managers were required to complete a questionnaire on personal assessment of calving procedures, neonate management and environmental factors. Results were collated and matched with the calf mortality rate of 2006 determined from the HI-Tier database for each farm. Randomly selected serum samples of a mean number of 7 calves at the age 6 months per herd were investigated for antibodies against bovine respiratory syncytial virus (BRSV-AB) and parainfluenzavirus type 3 (PIV3-AB). According to the proportion of calves with BRSV-AB or PIV3-AB (≤ 20 % or > 20 %) farms were divided into 2 groups. RESULTS: Customary timing of the first colostrum feeding as well as the perceived level of importance of EBP to the farm manager, as described in the questionnaire, showed a positive correlation to calf mortality. BRSV-AB occurred more frequently on farms where managers stated that the first colostrum feeding occurred later than 4 hours after birth, that birth monitoring was rarely practiced and that the estimated level of dust in the calf barn was considered high. PIV3-AB was more frequently found at farms practicing tethered calving. CONCLUSION AND CLINICAL RELEVANCE: The results of this study indicate that peri- and postnatal calf management procedures may affect calf mortality and the frequency of occurrence of BRSV-AB or PIV3-AB. The influences of birth monitoring and the time of first colostrum feeding as well as dust exposure should be taken into account in future studies on the frequency of EBP and be included in the veterinary cause analysis of herd EBP-related problems.


Assuntos
Anticorpos Antivirais/sangue , Pneumonia Enzoótica dos Bezerros , Animais , Animais Recém-Nascidos , Bovinos , Colostro , Feminino , Vírus da Parainfluenza 3 Humana/imunologia , Pneumonia Enzoótica dos Bezerros/epidemiologia , Pneumonia Enzoótica dos Bezerros/mortalidade , Gravidez , Vírus Sincicial Respiratório Bovino/imunologia , Fatores de Risco
13.
Antiviral Res ; 162: 54-60, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30550799

RESUMO

Human parainfluenza virus 3 (PIV3) and respiratory syncytial virus (RSV) are major causative agents of serious respiratory tract illness in newborns and infants. Maternal vaccination could be a promising approach to provide immediate protection against severe PIV3 and RSV infection in young infants. Previously, we demonstrated that maternal immunization with a subunit vaccine consisting of the RSV fusion (F) protein formulated with TriAdj, an adjuvant consisting of poly(I:C), immune defense regulatory peptide and polyphosphazene, protects newborn lambs from RSV. In the present study we evaluated the protective efficacy of a novel bivalent RSV-PIV3 vaccine candidate, FRipScHN/TriAdj, as a maternal vaccine against PIV3 infection in a neonatal lamb model. This vaccine consists of the pre-fusion form of the RSV F protein linked to the haemagglutinin-neuraminidase (HN) of PIV3, formulated with TriAdj. First, we successfully established PIV3 infection in neonatal lambs. Lambs infected with human PIV3 showed gross pathology, bronchointerstitial pneumonia and viral replication in the lungs. Subsequently, ewes were immunized with FRipScHN/TriAdj. RSV FRipSc- and PIV3 HN-specific antibodies with virus-neutralizing activity were detected in both the serum and the colostrum of the vaccinated ewes. The newborn lambs had RSV- and PIV3- neutralizing antibodies in their serum, which demonstrates that maternal antibodies were transferred to the neonates. At three days of age, the newborn lambs received an intrapulmonary challenge with PIV3. The lung pathology and virus production were significantly reduced in lambs that had received PIV3-specific maternal antibodies compared to lambs born to non-vaccinated ewes. These results suggest that maternal vaccination with a bivalent FRipScHN/TriAdj vaccine might be an effective method to provide protection against both PIV3 and RSV in neonates.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Anticorpos Antivirais/sangue , Imunidade Materno-Adquirida , Vacinas contra Parainfluenza/administração & dosagem , Infecções por Respirovirus/veterinária , Animais , Animais Recém-Nascidos , Anticorpos Neutralizantes/sangue , Feminino , Glicoproteínas/genética , Glicoproteínas/imunologia , Humanos , Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Gravidez , Vírus Sinciciais Respiratórios/genética , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/prevenção & controle , Ovinos , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Proteínas Virais de Fusão/administração & dosagem , Proteínas Virais de Fusão/imunologia
14.
Curr Pharm Biotechnol ; 19(11): 896-901, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30381069

RESUMO

BACKGROUND: Bovine Parainfluenza Virus type 3 (BPIV3) is a major but often overlooked pathogen that causes respiratory disease in cattle, especially during transportation and in feedlot situations. There is a demand for the rapid detection and serological diagnosis of BPIV3 to monitor the presence of the virus and its antibodies in cattle, which is critical in designing suitable interventions and control. METHODS: In the present study, ssDNA aptamers with high affinity and specificity against the HN protein of BPIV3 were selected using microplates as the matrix. RESULTS: After eleven rounds selection, thirty-four different DNA sequences were obtained in total, wherein w-32, w-33, and w-34 were repeated seven, eleven, and nine times, and with Kd values of 56.57 ± 2.7 nM, 24.64 ± 2.84 nM, and 31.3 ± 3.32 nM, respectively. Two-dimensional structural analysis showed that the three aptamers had several loop structures that were probably more energetically favorable for target binding. Of the three candidates, aptamer w-33 showed the best affinity in an indirect enzyme-linked aptamer assay (ELAA). The percent inhibition cutoff value of the ELAA, assessed using twenty negative sera, was 31%. CONCLUSION: In a comparative study with commercial ELISA kits, the positive detection rate of the ELAA was slightly higher than that of the commercial ELISA kits, and the coincidence rate of ELAA and ELISA was 88%. Further optimization of the ELAA method with more serums is needed.


Assuntos
Anticorpos Antivirais/sangue , Aptâmeros de Nucleotídeos/genética , DNA de Cadeia Simples/genética , Proteína HN/genética , Vírus da Parainfluenza 3 Humana/imunologia , Técnica de Seleção de Aptâmeros/métodos , Animais , Sequência de Bases , Bovinos , Ensaio de Imunoadsorção Enzimática/métodos , Vírus da Parainfluenza 3 Humana/genética , Técnica de Seleção de Aptâmeros/veterinária , Sensibilidade e Especificidade
15.
Proc Natl Acad Sci U S A ; 115(48): 12265-12270, 2018 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-30420505

RESUMO

Parainfluenza virus types 1-4 (PIV1-4) are highly infectious human pathogens, of which PIV3 is most commonly responsible for severe respiratory illness in newborns, elderly, and immunocompromised individuals. To obtain a vaccine effective against all four PIV types, we engineered mutations in each of the four PIV fusion (F) glycoproteins to stabilize their metastable prefusion states, as such stabilization had previously enabled the elicitation of high-titer neutralizing antibodies against the related respiratory syncytial virus. A cryoelectron microscopy structure of an engineered PIV3 F prefusion-stabilized trimer, bound to the prefusion-specific antibody PIA174, revealed atomic-level details for how introduced mutations improved stability as well as how a single PIA174 antibody recognized the trimeric apex of prefusion PIV3 F. Nine combinations of six newly identified disulfides and two cavity-filling mutations stabilized the prefusion PIV3 F immunogens and induced 200- to 500-fold higher neutralizing titers in mice than were elicited by PIV3 F in the postfusion conformation. For PIV1, PIV2, and PIV4, we also obtained stabilized prefusion Fs, for which prefusion versus postfusion titers were 2- to 20-fold higher. Elicited murine responses were PIV type-specific, with little cross-neutralization of other PIVs. In nonhuman primates (NHPs), quadrivalent immunization with prefusion-stabilized Fs from PIV1-4 consistently induced potent neutralizing responses against all four PIVs. For PIV3, the average elicited NHP titer from the quadrivalent immunization was more than fivefold higher than any titer observed in a cohort of over 100 human adults, highlighting the ability of a prefusion-stabilized immunogen to elicit especially potent neutralization.


Assuntos
Vírus da Parainfluenza 1 Humana/imunologia , Vírus da Parainfluenza 2 Humana/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Vírus da Parainfluenza 4 Humana/imunologia , Infecções por Respirovirus/imunologia , Proteínas Virais de Fusão/química , Vacinas Virais/química , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Microscopia Crioeletrônica , Feminino , Humanos , Macaca mulatta , Masculino , Camundongos , Vírus da Parainfluenza 1 Humana/química , Vírus da Parainfluenza 1 Humana/genética , Vírus da Parainfluenza 2 Humana/química , Vírus da Parainfluenza 2 Humana/genética , Vírus da Parainfluenza 3 Humana/química , Vírus da Parainfluenza 3 Humana/genética , Vírus da Parainfluenza 4 Humana/química , Vírus da Parainfluenza 4 Humana/genética , Infecções por Vírus Respiratório Sincicial , Infecções por Respirovirus/prevenção & controle , Infecções por Respirovirus/virologia , Proteínas Virais de Fusão/administração & dosagem , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Vacinas Virais/imunologia
16.
Antiviral Res ; 158: 78-87, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30071204

RESUMO

Human respiratory syncytial virus (RSV) and parainfluenza virus type 3 (PIV3) are major causes of serious lower respiratory tract disease in infants. Currently there is no licensed vaccine against RSV or PIV3. To make an effective bivalent subunit vaccine, a chimeric truncated FRHN protein containing the N-terminal ectodomain of the RSV fusion (F) protein linked to the C-terminal ectodomain of the PIV3 haemagglutinin-neuraminidase (HN) protein was produced in HEK293T cells. Mice, cotton rats and hamsters were immunized intramuscularly (IM) with both RSV F and PIV3 HN (FR+HN) or FRHN, formulated with TriAdj, which consists of poly(I:C), innate defense regulator peptide and poly[di(sodium carboxylatoethylphenoxy)]-phosphazene. Both formulations were immunogenic and elicited full protection from RSV; however, animals vaccinated with FRHN/TriAdj were significantly better protected from PIV3 than animals vaccinated with FR+HN/TriAdj. To develop a potentially more effective subunit vaccine, a chimeric glycoprotein (FRipScHN), encoding the RSV F ectodomain stabilized in the pre-fusion form linked to PIV3 HN was generated. Intramuscular vaccination with FRipScHN/TriAdj induced virus neutralizing antibodies followed by complete protection from RSV and PIV3 replication in the lungs of challenged cotton rats. Furthermore, intranasal vaccination with FRipScHN/TriAdj significantly reduced both RSV and PIV3 replication in cotton rats. Mucosal immunization with FRipScHN/TriAdj also elicited strong antigen-specific mucosal and systemic immune responses in a lamb model. In conclusion, the chimeric FRipScHN protein combined with TriAdj has potential for development of a safe, effective, bivalent vaccine against both RSV and PIV3.


Assuntos
Adjuvantes Imunológicos/farmacologia , Glicoproteínas/farmacologia , Vírus da Parainfluenza 3 Humana/imunologia , Substâncias Protetoras/farmacologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sincicial Respiratório Humano/imunologia , Infecções por Respirovirus/prevenção & controle , Animais , Proteínas Quimerinas/imunologia , Chlorocebus aethiops , Cricetinae , Modelos Animais de Doenças , Células HEK293 , Humanos , Imunidade Inata , Imunidade nas Mucosas , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Poli I-C , Infecções por Vírus Respiratório Sincicial/imunologia , Vacinas contra Vírus Sincicial Respiratório/imunologia , Infecções por Respirovirus/imunologia , Ovinos , Sigmodontinae , Vacinação , Vacinas de Subunidades Antigênicas , Proteínas Virais de Fusão/imunologia , Replicação Viral/efeitos dos fármacos
17.
Vaccine ; 35(51): 7139-7146, 2017 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-29153777

RESUMO

Human parainfluenza virus type 3 (PIV3) is a major cause of lower respiratory disease i.e. bronchitis, bronchiolitis or pneumonia, in infants and young children. Presently there is no licensed vaccine against PIV3. To produce an effective subunit vaccine, a chimeric FHN glycoprotein consisting of the N-terminal ectodomain of the fusion (F) protein linked to the haemagglutinin-neuraminidase (HN) protein without transmembrane domain, and secreted forms of the individual F and HN glycoproteins, were expressed in mammalian cells and purified. Mice and cotton rats were immunized intramuscularly (IM) with FHN or both F and HN proteins (F + HN), formulated with poly(I:C) and an innate defense regulator peptide in polyphosphazene (TriAdj). Significantly higher levels of systemic virus-neutralizing antibodies were observed in mice and cotton rats immunized with FHN/TriAdj when compared to animals immunized with the combination of F and HN proteins (F + HN/TriAdj). As PIV3 is a pneumotropic virus, another goal is to produce an effective mucosal subunit vaccine. Intranasal (IN) administration with FHN/TriAdj resulted in mucosal IgA production in the lung and virus neutralizing antibodies in the sera. After PIV3 challenge no virus was detected in cotton rats immunized with FHN/TriAdj regardless of the route of delivery. Protective immunity against PIV3 was also induced by FHN/TriAdj in hamsters. In conclusion, the FHN protein formulated with TriAdj has potential for development of a safe and effective vaccine against PIV3.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Proteína HN/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Proteínas Virais de Fusão/imunologia , Vacinas Virais/imunologia , Adjuvantes Imunológicos/química , Animais , Anticorpos Neutralizantes/biossíntese , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Cricetinae , Proteína HN/administração & dosagem , Proteína HN/genética , Humanos , Imunização , Camundongos , Poli I-C/administração & dosagem , Poli I-C/imunologia , Polilisina/administração & dosagem , Polilisina/imunologia , Sigmodontinae , Vacinação , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/efeitos adversos , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia , Proteínas Virais de Fusão/administração & dosagem , Proteínas Virais de Fusão/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/efeitos adversos
18.
Innate Immun ; 23(2): 147-154, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27913791

RESUMO

BMAL1 (brain and muscle ARNT-like protein 1, also known as MOP3 or ARNT3) belongs to the family of the basic helix-loop-helix (bHLH)-PAS domain-containing transcription factors, and is a key component of the molecular oscillator that generates circadian rhythms. Here, we report that BMAL1-deficient cells are significantly more susceptible to infection by two major respiratory viruses of the Paramyxoviridae family, namely RSV and PIV3. Embryonic fibroblasts from Bmal1-/- mice produced nearly 10-fold more progeny virus than their wild type controls. These results were supported by animal studies whereby pulmonary infection of RSV produced a more severe disease and morbidity in Bmal1-/-mice. These results show that BMAL1 can regulate cellular innate immunity against specific RNA viruses.


Assuntos
Fatores de Transcrição ARNTL/metabolismo , Fibroblastos/imunologia , Pulmão/patologia , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Infecções por Respirovirus/imunologia , Fatores de Transcrição ARNTL/genética , Animais , Linhagem Celular , Relógios Circadianos/genética , Fibroblastos/virologia , Humanos , Imunidade Inata/genética , Camundongos , Camundongos Knockout , RNA Interferente Pequeno/genética
19.
Cytotherapy ; 18(12): 1515-1524, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27692559

RESUMO

BACKGROUND AIMS: Human parainfluenza virus-3 (HPIV) is a common cause of respiratory infection in immunocompromised patients and currently has no effective therapies. Virus-specific T-cell therapy has been successful for the treatment or prevention of viral infections in immunocompromised patients but requires determination of T-cell antigens on targeted viruses. METHODS: HPIV3-specific T cells were expanded from peripheral blood of healthy donors using a rapid generation protocol targeting four HPIV3 proteins. Immunophenotyping was performed by flow cytometry. Viral specificity was determined by interferon (IFN)-γ ELISpot, intracellular cytokine staining and cytokine measurements from culture supernatants by Luminex assay. Cytotoxic activity was tested by 51Cr release and CD107a mobilization assays. Virus-specific T cells targeting six viruses were then produced by rapid protocol, and the phenotype of HPIV3-specific T cells was determined by immunomagnetic sorting for IFN-γ-producing cells. RESULTS: HPIV3-specific T cells were expanded from 13 healthy donors. HPIV3-specific T cells showed a CD4+ predominance (mean CD4:CD8 ratio 2.89) and demonstrated specificity for multiple HPIV3 antigens. The expanded T cells were polyfunctional based on cytokine production but only had a minor cytotoxic component. T cells targeting six viruses in a single product similarly showed HPIV3 specificity, with a predominant effector memory phenotype (CD3+/CD45RA-/CCR7-) in responder cells. DISCUSSION: HPIV3-specific T cells can be produced using a rapid ex vivo protocol from healthy donors and are predominantly CD4+ T cells with Th1 activity. HPIV3 epitopes can also be successfully targeted alongside multiple other viral epitopes in production of six-virus T cells, without loss of HPIV3 specificity. These products may be clinically beneficial to combat HPIV3 infections by adoptive T-cell therapy in immune-compromised patients.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Respirovirus/terapia , Relação CD4-CD8 , Células Cultivadas , Citometria de Fluxo , Humanos , Hospedeiro Imunocomprometido , Imunofenotipagem , Interferon gama/imunologia , Contagem de Linfócitos , Infecções por Respirovirus/imunologia
20.
J Virol ; 90(24): 11145-11156, 2016 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-27707917

RESUMO

A major arm of cellular innate immunity is type I interferon (IFN), represented by IFN-α and IFN-ß. Type I IFN transcriptionally induces a large number of cellular genes, collectively known as IFN-stimulated gene (ISG) proteins, which act as antivirals. The IFIT (interferon-induced proteins with tetratricopeptide repeats) family proteins constitute a major subclass of ISG proteins and are characterized by multiple tetratricopeptide repeats (TPRs). In this study, we have interrogated IFIT proteins for the ability to inhibit the growth of human parainfluenza virus type 3 (PIV3), a nonsegmented negative-strand RNA virus of the Paramyxoviridae family and a major cause of respiratory disease in children. We found that IFIT1 significantly inhibited PIV3, whereas IFIT2, IFIT3, and IFIT5 were less effective or not at all. In further screening a set of ISG proteins we discovered that several other such proteins also inhibited PIV3, including IFITM1, IDO (indoleamine 2,3-dioxygenase), PKR (protein kinase, RNA activated), and viperin (virus inhibitory protein, endoplasmic reticulum associated, interferon inducible)/Cig5. The antiviral effect of IDO, the enzyme that catalyzes the first step of tryptophan degradation, could be counteracted by tryptophan. These results advance our knowledge of diverse ISG proteins functioning as antivirals and may provide novel approaches against PIV3. IMPORTANCE: The innate immunity of the host, typified by interferon (IFN), is a major antiviral defense. IFN inhibits virus growth by inducing a large number of IFN-stimulated gene (ISG) proteins, several of which have been shown to have specific antiviral functions. Parainfluenza virus type 3 (PIV3) is major pathogen of children, and no reliable vaccine or specific antiviral against it currently exists. In this article, we report several ISG proteins that strongly inhibit PIV3 growth, the use of which may allow a better antiviral regimen targeting PIV3.


Assuntos
Proteínas de Transporte/imunologia , Interações Hospedeiro-Patógeno , Imunidade Inata , Interferon-alfa/imunologia , Interferon beta/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Células A549 , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/imunologia , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/virologia , Regulação da Expressão Gênica , Células HEK293 , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Interferon-alfa/genética , Interferon beta/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Vírus da Parainfluenza 3 Humana/crescimento & desenvolvimento , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Proteínas/genética , Proteínas/imunologia , Proteínas de Ligação a RNA , Transdução de Sinais , Triptofano/farmacologia , eIF-2 Quinase/genética , eIF-2 Quinase/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...