RESUMO
Peste des petits ruminants virus (PPRV) is a Morbillivirus that causes highly contagious and severe disease in various ruminants. PPRV infection leads to a severe inhibition of host antiviral immune response. Our previous study demonstrated that PPRV V protein blocks IFN response by targeting STAT proteins. In the current study, we identified the phosphoprotein (P) as a novel antagonistic factor of PPRV to counteract host antiviral innate immune response. PPRV P protein significantly suppressed RIG-I-like receptor pathway signaling and impaired IFN-ß and ISGs expression by targeting IFN regulatory factor (IRF)3 in both human embryonic kidney 293T cells and primary goat fibroblasts. The 1-102 region of P protein was critical for the antagonistic function of P protein. P protein interacted with IRF association domain (IAD) of IRF3 to block the interaction between TBK1 and IRF3. The interaction between TBK1 and the IAD of IRF3 is responsible for triggering the phosphorylation of IRF3. P protein competed with TBK1 to bind to the IAD of IRF3 that contributed to the decreased phosphorylation of IRF3, which, in turn, interfered with the dimerization of IRF3 and blocked IRF3 nuclear transportation. Besides, we also found that P protein interacted with IRF5 and IRF8. However, the involved mechanism remains unknown. Taken together, our results reveal a novel mechanism by which PPRV P protein antagonizes host antiviral innate immune response by interacting with the transcription factor IRF3, thereby inhibiting the type I IFN production and promoting viral replication.
Assuntos
Proteína DEAD-box 58/metabolismo , Fibroblastos/fisiologia , Fator Regulador 3 de Interferon/metabolismo , Peste dos Pequenos Ruminantes/imunologia , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Fosfoproteínas/metabolismo , Proteínas Virais/metabolismo , Animais , Células Cultivadas , Cabras , Humanos , Evasão da Resposta Imune , Imunidade Inata , Fator Regulador 3 de Interferon/genética , Transdução de Sinais , Replicação ViralRESUMO
ETHNOPHARMACOLOGICAL RELEVANCE: Synthetic antiviral drugs have several limitations including high cost. Thus research on antiviral property of medicinal plants is continuously gaining importance. Polyalthia longifolia possesses several medicinal properties and has been used in traditional ayurvedic medicine for treatment of dermatological ailments as kushta, visarpa/herpes virus infection and also to treat pyrexia of unknown origin as mentioned in Visarpa Chikitsa. AIM OF THE STUDY: Keeping in view the cytotoxic, anti-cancer activity and antiviral efficacy of Polyalthia longifolia against herpes, present study was undertaken to evaluate the in vitro antiviral activity of methanolic extract of Polyalthia longifolia leaves, if any, and to unravel the possible target(s)/mechanism of action. MATERIAL AND METHODS: Antiviral activity of Polyalthia longifolia methanolic extract was studied using Vero cell lines against paramyxoviruses, namely-peste des petits ruminants virus (PPRV) and Newcastle disease virus (NDV). Cytotoxicity of the test extract was evaluated employing MTT assay. Virucidal activity, and viral-attachment, virus entry and release assays were determined in Vero cells using standard experimental protocols. The viral RNA in the virus-infected cells was quantified by qRT-PCR. RESULTS: At non-cytotoxic concentration, methanolic extract of Polyalthia longifolia leaves was found to inhibit the replication of PPRV and NDV at viral entry and budding level, whereas other steps of viral life cycle such as attachment and RNA synthesis remained unaffected. CONCLUSIONS: Polyalthia longifolia leaves extract possesses promising antiviral activity against paramyxoviruses and acts by inhibiting the entry and budding of viruses; and this plant extract evidently possesses excellent and promising potential for development of effective herbal antiviral drug.
Assuntos
Antivirais/farmacologia , Vírus da Doença de Newcastle/efeitos dos fármacos , Vírus da Peste dos Pequenos Ruminantes/efeitos dos fármacos , Extratos Vegetais/farmacologia , Polyalthia , Animais , Chlorocebus aethiops , Vírus da Doença de Newcastle/fisiologia , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Folhas de Planta , Células Vero , Internalização do Vírus/efeitos dos fármacosRESUMO
Peste-des-petits-ruminants virus (PPRV) causes a severe respiratory disease in small ruminants. The possible impact of different atypical host species in the spread and planed worldwide eradication of PPRV remains to be clarified. Recent transmission trials with the virulent PPRV lineage IV (LIV)-strain Kurdistan/2011 revealed that pigs and wild boar are possible sources of PPRV-infection. We therefore investigated the role of cattle, llamas, alpacas, and dromedary camels in transmission trials using the Kurdistan/2011 strain for intranasal infection and integrated a literature review for a proper evaluation of their host traits and role in PPRV-transmission. Cattle and camelids developed no clinical signs, no viremia, shed no or only low PPRV-RNA loads in swab samples and did not transmit any PPRV to the contact animals. The distribution of PPRV-RNA or antigen in lymphoid organs was similar in cattle and camelids although generally lower compared to suids and small ruminants. In the typical small ruminant hosts, the tissue tropism, pathogenesis and disease expression after PPRV-infection is associated with infection of immune and epithelial cells via SLAM and nectin-4 receptors, respectively. We therefore suggest a different pathogenesis in cattle and camelids and both as dead-end hosts for PPRV.
Assuntos
Camelus/virologia , Especificidade de Hospedeiro , Interações Hospedeiro-Patógeno , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Biomarcadores , Biópsia , Bovinos , Feminino , Testes Hematológicos , Imuno-Histoquímica , Masculino , Peste dos Pequenos Ruminantes/sangue , Peste dos Pequenos Ruminantes/patologiaRESUMO
The measles virus (MeV), a member of the genus Morbillivirus, is an established pathogen of humans. A key feature of morbilliviruses is their ability to spread by virus-cell and cell-cell fusion. The latter process, which leads to syncytia formation in vitro and in vivo, is driven by the viral fusion (F) and haemagglutinin (H) glycoproteins. In this study, we demonstrate that MeV glycoproteins are sensitive to inhibition by bone marrow stromal antigen 2 (BST2/Tetherin/CD317) proteins. BST2 overexpression causes a large reduction in MeV syncytia expansion. Using quantitative cell-cell fusion assays, immunolabeling, and biochemistry we further demonstrate that ectopically expressed BST2 directly inhibits MeV cell-cell fusion. This restriction is mediated by the targeting of the MeV H glycoprotein, but not other MeV proteins. Using truncation mutants, we further establish that the C-terminal glycosyl-phosphatidylinositol (GPI) anchor of BST2 is required for the restriction of MeV replication in vitro and cell-cell fusion. By extending our study to the ruminant morbillivirus peste des petits ruminants virus (PPRV) and its natural host, sheep, we also confirm this is a broad and cross-species specific phenotype.
Assuntos
Antígenos CD/genética , Fusão Celular , Glicoproteínas/genética , Interações entre Hospedeiro e Microrganismos/genética , Vírus do Sarampo/genética , Vírus da Peste dos Pequenos Ruminantes/genética , Animais , Proteínas do Capsídeo/genética , Linhagem Celular , Células Epiteliais/virologia , Proteínas Ligadas por GPI/genética , Glicoproteínas/biossíntese , Células HEK293 , Humanos , Vírus do Sarampo/fisiologia , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Ovinos , Proteínas Virais de Fusão/genética , Replicação Viral/genéticaRESUMO
Peste des petits ruminants virus (PPRV), a member of the genus Morbillivirus, in the family Paramyxoviridae expresses two membrane glycoproteins, the fusion (F) and haemagglutinin (H) glycoproteins which mediate virus-to-cell fusion and cell-to-cell fusion leading to the induction of syncytia in PPRV infected cells. In the context of the characterization of the virulent lineage IV strain PPRV Kurdistan 2011, isolated from wild goats from the Kurdistan region in Iraq, we observed that both PPRV Kurdistan 2011 and the PPRV Nigeria 75/1 vaccine strain led to induction of large syncytia in Vero-dogSLAM cells within 48â¯h whereas both failed to induce detectable cell-cell fusion events in two Vero cell lines of differing passage histories. We were unable to detect syncytium formation in transiently transfected cells expressing PPRV F or H alone whereas co-expression of F and H induced large syncytia - in Vero-dogSLAM cells only. In VeroMontpellier cells expressing PPRV F and H, fused cells were rarely detectable indicating that PPRV mediated cell fusion activity is impaired in this cell line. Surprisingly, on Vero-dogSLAM cells the vaccine strain grew to titers of 105.25 TCID50/ml, whereas infectious virus yield was about 200-fold higher on VeroMontpellier and Vero-76 cells. In contrast, the virulent Kurdistan 2011 strain grew to a maximum titer of 107.0 TCID50/ml on Vero-dogSLAM cells and only 104.5 TCID50/ml on normal Vero cells. This was as expected since Vero cells lacking the SLAM receptor for PPRV are regarded as not so permissive for infection. To elucidate the divergent productive replication behaviour of PPRV Nigeria 75/1 vaccine strain on Vero vs Vero-dogSLAM cells, we examined whether intracellular transport and/or maturation of the viral envelope glycoproteins F and H might be implicated with this phenomenon. The results indicate that F in contrast to the H glycoprotein matures inefficiently during intracellular transport in VeroMontpellier cells, thus leading to an absence of detectable syncytia formation. However, in the case of the PPRV Nigeria 75/1 vaccine strain this did not impair efficient virus assembly and release.
Assuntos
Vírus da Peste dos Pequenos Ruminantes/fisiologia , Proteínas Virais de Fusão/metabolismo , Montagem de Vírus , Replicação Viral , Animais , Transporte Biológico , Chlorocebus aethiops , Doenças das Cabras/virologia , Cabras/virologia , Hemaglutininas Virais/metabolismo , Iraque , Peste dos Pequenos Ruminantes/prevenção & controle , Vírus da Peste dos Pequenos Ruminantes/classificação , Vírus da Peste dos Pequenos Ruminantes/imunologia , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Células VeroRESUMO
Peste des petits ruminants virus (PPRV) has emerged as a significant threat to the productivity of small ruminants worldwide. PPRV is lymphotropic in nature and induces in the hosts a transient but severe immunosuppression, especially innate immunity. However, it remains largely unknown how NK cells respond and are regulated at the earliest time points after an acute viral PPRV infection in goats. In this study, we revealed that multiple immune responses of goat peripheral NK cells were compromised during PPRV infection, including the cytolytic effector molecule expression and cytokine production. Importantly, we demonstrated that PPRV infection stimulated the expression of TWEAK, a negative regulator of cytotoxic function of NK cells, which may be involved in the suppression of cytotoxicity as well as cytokine production in infected goat NK cells. Furthermore, we found that PPRV infection induced TWEAK expression in goat NK cells involving post-transcription by suppressing miR-1, a novel negative miRNA directly targeting the TWEAK gene. Moreover, replication of virus is required for inhibition of miR-1 expression during PPRV infection, and the non-structural V protein of PPRV plays an important role in miR-1 mediated TWEAK upregulation. Additionally, we revealed that the regulation of NK cell immune responses by TWEAK is mediated by MyD88, SOCS1, and STAT3. Taken together, our results demonstrated that TWEAK may play a key role in regulating goat peripheral NK cell cytotoxicity and cytokine expression levels during PPRV infection.
Assuntos
Citocina TWEAK/genética , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , MicroRNAs/genética , Peste dos Pequenos Ruminantes/etiologia , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Fator de Necrose Tumoral alfa/genética , Animais , Regulação da Expressão Gênica , Cabras , Modelos Biológicos , Peste dos Pequenos Ruminantes/metabolismo , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Replicação ViralRESUMO
Peste des petits ruminants (PPR) is an acute and highly contagious disease in small ruminants that causes significant economic losses in developing countries. An increasing number of studies have demonstrated that both autophagy and apoptosis are important cellular mechanisms for maintaining homeostasis, and they participate in the host response to pathogens. However, the crosstalk between apoptosis and autophagy in host cells during PPRV infection has not been clarified. In this study, autophagy was induced upon virus infection in caprine endometrial epithelial cells (EECs), as determined by the appearance of double- and single-membrane autophagy-like vesicles, LC3-I/LC3-II conversion, and p62 degradation. We also found that PPRV infection triggered a complete autophagic response, most likely mediated by the non-structural protein C and nucleoprotein N. Moreover, our results suggest that autophagy not only promotes the replication of PPRV in EECs but also provides a potential mechanism for inhibiting PPRV-induced apoptosis. Inhibiting autophagosome formation by wortmannin and knocking down the essential autophagic proteins Beclin-1 and ATG7 induces caspase-dependent apoptosis in EECs in PPRV infection. However, inhibiting autophagosome and lysosome fusion by NH4Cl and chloroquine did not increase the number of apoptotic cells. Collectively, these data are the first to indicate that PPRV-induced autophagy inhibits caspase-dependent apoptosis and thus contributes to the enhancement of viral replication and maturity in host cells.
Assuntos
Apoptose , Autofagia , Caspases/metabolismo , Células Epiteliais/virologia , Vírus da Peste dos Pequenos Ruminantes/patogenicidade , Replicação Viral , Cloreto de Amônio/farmacologia , Androstadienos/farmacologia , Animais , Autofagossomos/efeitos dos fármacos , Proteína 7 Relacionada à Autofagia/deficiência , Proteína 7 Relacionada à Autofagia/genética , Proteína Beclina-1/deficiência , Proteína Beclina-1/genética , Cloroquina/farmacologia , Endométrio/citologia , Feminino , Cabras , Interações Hospedeiro-Patógeno , Humanos , Técnicas In Vitro , Lisossomos/efeitos dos fármacos , Lisossomos/fisiologia , Nucleoproteínas/metabolismo , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Proteína C/metabolismo , WortmaninaRESUMO
Peste des petits ruminants virus (PPRV) belongs to the genus Morbillivirus that causes an acute and highly contagious disease in goats and sheep. Virus infection can trigger the change in the cellular microRNA (miRNA) expression profile, which play important post-transcriptional regulatory roles in gene expression and can greatly influence viral replication and pathogenesis. Here, we employed deep sequencing technology to determine cellular miRNA expression profile in goat peripheral blood mononuclear cells (PBMC) infected with Nigeria 75/1 vaccine virus, a widely used vaccine strain for mass vaccination programs against Peste des petits ruminants. Expression analysis demonstrated that PPRV infection can elicit 316 significantly differentially expressed (DE) miRNA including 103 known and 213 novel miRNA candidates in infected PBMC at 24 hours post-infection (hpi) as compared with a mock control. Target prediction and functional analysis of these DEmiRNA revealed significant enrichment for several signaling pathways including TLR signaling pathways, PI3K-Akt, endocytosis, viral carcinogenesis, and JAK-STAT signaling pathways. This study provides a valuable basis for further investigation of the roles of miRNA in PPRV replication and pathogenesis.
Assuntos
Regulação da Expressão Gênica , Doenças das Cabras/genética , Leucócitos Mononucleares/metabolismo , MicroRNAs/genética , Peste dos Pequenos Ruminantes/genética , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Animais , China , Perfilação da Expressão Gênica/veterinária , Doenças das Cabras/virologia , Cabras , Sequenciamento de Nucleotídeos em Larga Escala/veterinária , MicroRNAs/metabolismo , Peste dos Pequenos Ruminantes/virologiaRESUMO
Peste des petits ruminants virus (PPRV), the etiological agent of peste des petits ruminants (PPR), causes an acute or subacute disease in small ruminants. Although abortion is observed in an unusually large proportion of pregnant goats during outbreaks of PPR, the pathogenic mechanism underlying remains unclear. Here, the gene expression profile of caprine endometrial epithelial cells (EECs) infected with PPRV Nigeria 75/1 was determined by DNA microarray to investigate the cellular response immediately after viral entry. The microarray analysis revealed that a total of 146 genes were significantly dysregulated by PPRV internalization within 1 h post-infection (hpi). Of these, 85 genes were upregulated and 61 genes were downregulated. Most of these genes, including NFKB1A, JUNB, and IL1A, have not previously been reported in association with PPRV infection in goats. Following viral replication (24 hpi), the expression of 307 genes were significantly upregulated and that of 261 genes were downregulated. The data for the genes differentially expressed in EECs were subjected to a time sequence profile analysis, gene network analysis and pathway analysis. The gene network analysis showed that 13 genes (EIF2AK3, IL10, TLR4, ZO3, NFKBIB, RAC1, HSP90AA1, SMAD7, ARG2, JUNB, ZFP36, APP, and IL1A) were located in the core of the network. We clearly demonstrate that PPRV infection upregulates the expression of nectin-4 after 1 hpi, which peaked at 24 hpi in EECs. In conclusion, this study demonstrates the early cellular gene expression in the caprine endometrial epithelial cells after the binding and entry of PPRV.
Assuntos
Endométrio/virologia , Cabras/genética , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Transcriptoma , Vírion/fisiologia , Ligação Viral , Animais , Endométrio/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Feminino , Cabras/virologia , Técnicas In VitroRESUMO
BACKGROUND: The specific and dynamic interaction between the hemagglutinin (H) and fusion (F) proteins of morbilliviruses is a prerequisite for the conformational rearrangements and membrane fusion during infection process. The two heptad repeat regions (HRA and HRB) of F protein are both important for the triggering of F protein. METHODS: In this study, the direct interactions of Peste des petits ruminants virus (PPRV) H with F, HRA and HRB were quantitatively evaluated using biosensor surface plasmon resonance (SPR). RESULTS: The binding affinities of immobilized pCMV-HA-H (HA-H) interacted with proteins pCMV-HA-F (HA-F) and pCMV-HA-HRB (HA-HRB) (KD = 1.91 × 10- 8 M and 2.60 × 10- 7 M, respectively) reacted an order of magnitude more strongly than that of pCMV-HA-HRA (HA-HRA) and pCMV-HA-Tp IGFR-LD (HA) (KD = 1.08 × 10- 4 M and 1.43 × 10- 4 M, respectively). CONCLUSIONS: The differences of the binding affinities suggested that HRB is involved in functionally important intermolecular interaction in the fusion process.
Assuntos
Hemaglutininas Virais/metabolismo , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Proteínas Virais de Fusão/metabolismo , Animais , Células CHO , Cricetulus , Citometria de Fluxo , Expressão Gênica , Hemaglutininas Virais/química , Hemaglutininas Virais/genética , Cinética , Ligação Proteica , Proteínas Recombinantes , Ressonância de Plasmônio de Superfície , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genéticaRESUMO
Sheep and goat pox (SGP) with peste des petits ruminants (PPR) are transboundary viral diseases of small ruminants that cause huge economic losses. Recombinant vaccines that can protect from both infections have been reported as a promising solution for the future. SGP was used as a vector to express two structural proteins hemagglutinin or the fusion protein of PPRV. We compared immunity conferred by recombinant capripoxvirus vaccines expressing H or F or both HF. Safety and efficacy were evaluated in goats and sheep. Two vaccine doses were tested in sheep, 104.5TCDI50 in 1ml dose was retained for the further experiment. Results showed that the recombinant HF confers an earlier and stronger immunity against both SGP and PPR. This recombinant vaccine protect also against the disease in exposed and unexposed sheep. The potential Differentiating Infected from Vaccinated Animals of recombinant vaccines is of great advantage in any eradication program.
Assuntos
Capripoxvirus/imunologia , Doenças das Cabras/prevenção & controle , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/imunologia , Infecções por Poxviridae/veterinária , Doenças dos Ovinos/prevenção & controle , Proteínas Virais/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/imunologia , Capripoxvirus/genética , Capripoxvirus/isolamento & purificação , Capripoxvirus/fisiologia , Doenças das Cabras/imunologia , Doenças das Cabras/virologia , Cabras , Hemaglutininas/administração & dosagem , Hemaglutininas/genética , Hemaglutininas/imunologia , Vírus da Peste dos Pequenos Ruminantes/genética , Vírus da Peste dos Pequenos Ruminantes/isolamento & purificação , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Infecções por Poxviridae/imunologia , Infecções por Poxviridae/prevenção & controle , Infecções por Poxviridae/virologia , Ovinos , Doenças dos Ovinos/imunologia , Doenças dos Ovinos/virologia , Proteínas Virais de Fusão/administração & dosagem , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia , Proteínas Virais/administração & dosagem , Proteínas Virais/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genéticaRESUMO
Peste des petits ruminants virus (PPRV), belonging to paramyxoviruses, has six structure proteins (such as matrix protein (M), nucleocapsid proteins (N), fusion protein (F) and hemagglutinin protein (H)) and could cause high morbidity and mortality in sheep and goats. Although a vaccine strain of PPRV has been rescued and co-expression of M and N could yield PPRV-like particles, the roles of structure proteins in virion assembly and release have not been investigated in detail. In this study, plasmids carrying PPRV cDNA sequences encoding the N, M, H, and F proteins were expressed in Vero cells. The co-expression of all four proteins resulted in the release of virus-like particles (VLPs) with similar release efficiency to that of authentic virions. Moreover, the co-expression of M together with F also resulted in efficient VLPs release. In the absence of M protein, the expression of no combination of the other proteins resulted in particle release. In summary, a VLPs production system for PPRV has been established and M protein is necessary for promoting the assembly and release of VLPs, of which the predominant protein is M protein. Further study will be focused on the immunogenicity of the VLPs.
Assuntos
Vírus da Peste dos Pequenos Ruminantes/metabolismo , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Células Vero/metabolismo , Proteínas da Matriz Viral/metabolismo , Animais , Anticorpos Antivirais , Chlorocebus aethiops/metabolismo , Chlorocebus aethiops/fisiologia , DNA Complementar , DNA Viral , Hemaglutininas Virais/metabolismo , Hemaglutininas Virais/fisiologia , Camundongos , Proteínas do Nucleocapsídeo/metabolismo , Proteínas do Nucleocapsídeo/fisiologia , Vírus da Peste dos Pequenos Ruminantes/genética , Vírus da Peste dos Pequenos Ruminantes/imunologia , Proteínas Virais de Fusão/metabolismo , Proteínas Virais de Fusão/fisiologiaRESUMO
In this study, we found out that blocking the receptor tyrosine kinase (RTK) signaling in Vero cells by tryphostin AG879 impairs the in vitro replication of the peste des petits ruminants virus (PPRV). A reduced virus replication in Trk1-knockdown (siRNA) Vero cells confirmed the essential role of RTK in the virus replication, in particular a specific regulation of viral RNA synthesis. These data represent the first evidence that the RTK signaling regulates replication of a morbillivirus.
Assuntos
Peste dos Pequenos Ruminantes/enzimologia , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Receptor trkA/metabolismo , Transdução de Sinais , Replicação Viral , Animais , Chlorocebus aethiops , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/genética , Receptor trkA/genética , Células VeroRESUMO
Small ruminants infected with peste des petits ruminants virus exhibit lesions typical of epithelial infection and necrosis. However, the only established host receptor for this virus is the immune cell marker signaling lymphocyte activation molecule (SLAM). We have confirmed that the ovine Nectin-4 protein, when overexpressed in epithelial cells, permits efficient replication of PPRV. Furthermore, this gene was predominantly expressed in epithelial tissues and encoded by multiple haplotypes in sheep breeds from around the world.
Assuntos
Moléculas de Adesão Celular/metabolismo , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Receptores Virais/metabolismo , Ligação Viral , Animais , Células Epiteliais/virologia , Nectinas , Vírus da Peste dos Pequenos Ruminantes/crescimento & desenvolvimento , OvinosRESUMO
The lung and gut morphometry in both natural and experimental Peste de petit ruminant (PPR) virus which are scanty in literature hence the need for this study. The goats that were submitted for necropsy in the Department of Veterinary Pathology University of Ibadan between 2009 and 2010 and the gross pathological diagnosis were PPR were enrolled in this study. The degree of pneumonia as a percentage of the total lung volume was estimated using standard methods. The gut morphometry of goats experimentally infected with PPR virus was also used. Student "T" test was used for the test of significance in evaluating the effect of age, sex and the lung consolidation pattern in natural PPR and analysis of the gut morphometry. Complicated PPR had significant higher pulmonary consolidation when compared with the uncomplicated PPR (p< 0.05). The pulmonary consolidation was significantly higher on the right lung with a mean percentage value of 6.54 than the left lung (p< 0.05). The caudal lobe was more consolidated than the cranial and middle lobes in natural PPR. The pulmonary consolidation was more in goats less than a year, while the buck had a significantly higher pulmonary consolidation than the does (p< 0.05). There was no significant difference in the mean length of the villi and width of the villi of PPR virus infected goats when compared to the control, however a significant difference was observed in the cryptal depth (p< 0.05). There was a significant difference in the mean villi length and cryptal depth of goats with complicated PPR (Mannheimia hemolytica) infected goats (p< 0.05) relative to the control. From this study, it showed that most natural PPR were complicated with bacteria and this complication may have contributed to the fatality associated with PPR especially those caused by lineage 1 viruses. This study also showed that secondary bacterial involvement in course of PPR affect the gut morphometry and that could account for the severity of intestinal lesion commonly observed with field PPR in Nigerian goats.
La morfometría del pulmón y el intestino en la infección del virus Peste des petits ruminants (PRR) de forma natural así como experimental es escaza en la literatura, de ahí la necesidad de este estudio. Fueron incluidas en este estudio las cabras que fueron sometidas a autopsia en el Departamento de Patología Veterinaria de la Universidad de Ibadan entre 2009 y 2010, con diagnóstico patológico macroscópico de PPR. El grado de neumonía como porcentaje del volumen pulmonar total fue estimado mediante los métodos estándar. También fue determinada la morfometría del intestino de las cabras infectadas experimentalmente con el virus PPR. Se utilizó la prueba "T" de Student para determinar la significancia en la evaluación de los efectos de edad, sexo, patrón de consolidación pulmonar en PPR natural y análisis de la morfometría intestinal. La PPR complicada tuvo una consolidación pulmonar altamente significativa en comparación con la PPR no complicada (p <0,05). La consolidación pulmonar fue significativamente mayor en el pulmón derecho, con un valor porcentaje promedio de 6,54 en comparación al pulmón izquierdo (p <0,05). El lóbulo caudal fue más consolidado que los lóbulos craneal y medio en presencia del PPR natural. La consolidación pulmonar fue más frecuente en caprinos menores de un año, mientras que los machos cabríos tuvieron una consolidación pulmonar significativamente más alta (p <0,05). No hubo diferencias significativas en la longitud y ancho promedio de las vellosidades en cabras infectadas con PPR en comparación con el control, pero se observó una diferencia significativa en la profundidad de las criptas (p <0,05). Hubo diferencia significativa en la longitud de las vellosidades y la profundidad media de las criptas en las cabras infectadas con PPR complicada (Mannheimia haemolytica) (p <0,05) en relación al control. A partir de este estudio, se demostró que las infecciones con PPR natural se complicaron con bacterias, y estas complicaciones pueden haber contribuido a la mortalidad asociada el PPR, especialmente las causadas por el virus del linaje 1. Este estudio también mostró que la participación bacteriana secundaria en el curso de la PPR afecta la morfometría intestinal y que podría dar cuenta de la gravedad de la lesión intestinal observada comúnmente en la infección de PPR en cabras de Nigeria.
Assuntos
Animais , Doenças das Cabras/virologia , Peste dos Pequenos Ruminantes/virologia , Trato Gastrointestinal/patologia , Fígado/patologia , Cabras , Fatores Sexuais , Fatores Etários , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Peste dos Pequenos Ruminantes/patologia , Trato Gastrointestinal/virologia , Fígado/virologiaRESUMO
A specific interaction between the F and H proteins is required to enable fusion of the virus and host cell membranes and in some cases these proteins are not interchangeable between related viruses of the family Paramyxoviridae. For example, the F and H proteins of two ruminant morbilliviruses, rinderpest virus (RPV) and Peste-des-petits-ruminants virus (PPRV), are not interchangeable since viable virus could not be rescued from cDNA constructs where an individual glycoprotein gene of RPV was replaced with that from PPRV. To investigate which domain of the H protein, extracellular or cytoplasmic/transmembrane, was most important for preventing this interaction, two chimeric H gene constructs were made where the normal H gene of RPV was substituted with variant H genes where the transmembrane/cytoplasmic tail region (pRPV2C-PPRTm) or the whole ectodomain (pRPV2C-PPRExt) were derived from PPRV. Chimeric viruses were rescued from both the constructs and, while RPV2C-PPRTm virus grew to as high titres as the parent virus, RPV2C-PPRExt virus was extremely debilitated with respect to growth in tissue culture. Thus the ectodomain of H is the most important region required for effective interactions of the two glycoproteins for the recovery of viable virus. Nevertheless, the transmembrane/cytoplasmic domain of RPV alone can allow a chimeric virus to be rescued, which was not possible when the complete H gene was derived from PPRV. Both versions of the H protein and also the F protein were found to be incorporated into the envelope of the budded virions.
Assuntos
Hemaglutininas Virais/química , Hemaglutininas Virais/fisiologia , Vírus da Peste Bovina/fisiologia , Replicação Viral/genética , Animais , Chlorocebus aethiops , Citoplasma , DNA Complementar , Expressão Gênica , Glicoproteínas/metabolismo , Hemaglutininas Virais/genética , Proteínas de Membrana , Microscopia de Fluorescência , Microscopia Imunoeletrônica , Vírus da Peste dos Pequenos Ruminantes/genética , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Estrutura Terciária de Proteína/genética , RNA Mensageiro/análise , RNA Viral/análise , Proteínas Recombinantes de Fusão/metabolismo , Vírus da Peste Bovina/genética , Células Vero , Proteínas Virais de Fusão/metabolismo , Ensaio de Placa Viral , Proteínas Virais/análiseRESUMO
We describe the properties of the two heptad repeats (HR1 and HR2) of the Peste des petits ruminants virus (PPRV) fusion protein (F) to obtain insights into the mechanism by which these repeats influence PPRV-mediated cell fusion. Both HR1 and HR2 inhibit PPRV-mediated syncytia formation in Vero cells in vitro. Of these, HR2 was found to be more effective than HR1. We studied the mechanism of fusion inhibition by these two repeats by using various biophysical and biochemical methods either separately or together. CD spectral analysis of these repeats revealed that the alpha-helical content of HR1 and HR2 when used together is higher than that of their simulated spectrum in the mixture, suggesting the formation of a highly structured complex by these repeats. Protease protection assays confirmed that such a complex is highly stable. Electrospray mass spectrometry of protease-digested products of the HR1-HR2 complex showed protection of fragments corresponding to both HR1 and HR2 sequences involved in complex formation. By employing size-exclusion chromatography and chemical cross-linking experiments, we show that three units each of HR1 and HR2 form a complex in which HR1 is a trimer and HR2 is a monomer. Homology-based three-dimensional modeling of this complex showed that HR1 and HR2 together form a six-helix and trimeric coiled-coil bundle. In this model, the HR1 trimer forms the core whereas HR2, while interacting with HR1 in an antiparallel orientation, forms a two-stranded coiled-coil structure and lies at the periphery of the structure. These results are discussed in the context of a common fusion mechanism among paramyxoviruses.
Assuntos
Glicoproteínas/química , Glicoproteínas/fisiologia , Vírus da Peste dos Pequenos Ruminantes/química , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/fisiologia , Montagem de Vírus/fisiologia , Motivos de Aminoácidos/genética , Motivos de Aminoácidos/fisiologia , Sequência de Aminoácidos , Animais , Antivirais/fisiologia , Chlorocebus aethiops , Endopeptidase K/metabolismo , Células Gigantes/fisiologia , Células Gigantes/virologia , Glicoproteínas/genética , Hidrólise , Proteínas de Membrana , Dados de Sequência Molecular , Vírus da Peste dos Pequenos Ruminantes/genética , Estrutura Secundária de Proteína/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Sequências Repetitivas de Aminoácidos/genética , Sequências Repetitivas de Aminoácidos/fisiologia , Vírus Sendai/química , Células Vero , Proteínas Virais de Fusão/genéticaRESUMO
To study the process of membrane fusion in Morbilliviruses, the fusion (F) glycoproteins of Peste des petits ruminants virus (PPRV) and Rinderpest virus (RPV) were expressed transiently in mammalian cells. The recombinant F proteins were found to be localized at the surface of transfected cells. The fusion activity, as evident from cell fusion assays and lysis of chicken erythrocytes, documented that transiently expressed PPRV F glycoprotein induces cell fusion in the absence of homotypic hemagglutinin-neuraminidase (HN) attachment glycoprotein. The coexpression of homotypic HN increased the extent of fusion by twofold, while the efficiency of fusion was found to be substantially enhanced. In contrast, in RPV F-expressing cells, fusion was detected only when homotypic hemagglutinin (H) or heterotypic HN protein was coexpressed. This differs from the strict type-specific requirement for the attachment protein as in the fusion process of most of the paramyxoviruses. Further, we demonstrate by fluorescence transfer experiments that while PPRV F brings about both hemifusion and complete fusion on its own, RPV F induces only hemifusion while it brings about complete fusion in the presence of homotypic or heterotypic attachment protein.
Assuntos
Proteína HN/fisiologia , Fusão de Membrana/fisiologia , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Proteínas Virais de Fusão/fisiologia , Animais , Linhagem Celular , Membrana Celular/metabolismo , Chlorocebus aethiops , Eritrócitos , Células Gigantes/fisiologia , Cobaias , Hemólise , Transfecção , Células Vero , Proteínas Virais de Fusão/genéticaRESUMO
There is evidence that CD46 (membrane cofactor protein) is a cellular receptor for vaccine and laboratory-passaged strains of measles virus (MV). Following infection with these MV strains, CD46 is downregulated from the cell surface, and consequent complement-mediated lysis has been shown to occur upon infection of a human monocytic cell line. The MV hemagglutinin (H) protein alone is capable of inducing this downregulation. Some wild-type strains of MV fail to downregulate CD46, despite infection being prevented by anti-CD46 antibodies. In this study we show that CD46 is also downregulated to the same extent by wild-type, vaccine, and laboratory-passaged strains of rinderpest virus (RPV), although CD46 did not appear to be the receptor for RPV. Expression of the RPV H protein by a nonreplicating adenovirus vector was also found to cause this downregulation. A vaccine strain of peste des petits ruminants virus caused slight downregulation of CD46 in infected Vero cells, while wild-type and vaccine strains of canine distemper virus and a wild-type strain of dolphin morbillivirus failed to downregulate CD46. Downregulation of CD46 can, therefore, be a function independent of the use of this protein as a virus receptor.