Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Virol ; 98(4): e0014624, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38440983

RESUMO

Peste des petits ruminants is an acute and highly contagious disease caused by the Peste des petits ruminants virus (PPRV). Host proteins play a crucial role in viral replication. However, the effect of fusion (F) protein-interacting partners on PPRV infection is poorly understood. In this study, we found that the expression of goat plasminogen activator urokinase (PLAU) gradually decreased in a time- and dose-dependent manner in PPRV-infected goat alveolar macrophages (GAMs). Goat PLAU was subsequently identified using co-immunoprecipitation and confocal microscopy as an F protein binding partner. The overexpression of goat PLAU inhibited PPRV growth and replication, whereas silencing goat PLAU promoted viral growth and replication. Additionally, we confirmed that goat PLAU interacted with a virus-induced signaling adapter (VISA) to antagonize F-mediated VISA degradation, increasing the production of type I interferon. We also found that goat PLAU reduced the inhibition of PPRV replication in VISA-knockdown GAMs. Our results show that the host protein PLAU inhibits the growth and replication of PPRV by VISA-triggering RIG-I-like receptors and provides insight into the host protein that antagonizes PPRV immunosuppression.IMPORTANCEThe role of host proteins that interact with Peste des petits ruminants virus (PPRV) fusion (F) protein in PPRV replication is poorly understood. This study confirmed that goat plasminogen activator urokinase (PLAU) interacts with the PPRV F protein. We further discovered that goat PLAU inhibited PPRV replication by enhancing virus-induced signaling adapter (VISA) expression and reducing the ability of the F protein to degrade VISA. These findings offer insights into host resistance to viral invasion and suggest new strategies and directions for developing PPR vaccines.


Assuntos
Doenças das Cabras , Cabras , Interações Hospedeiro-Patógeno , Peste dos Pequenos Ruminantes , Vírus da Peste dos Pequenos Ruminantes , Ativador de Plasminogênio Tipo Uroquinase , Proteínas Virais de Fusão , Animais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína DEAD-box 58/metabolismo , Doenças das Cabras/imunologia , Doenças das Cabras/metabolismo , Doenças das Cabras/virologia , Cabras/imunologia , Cabras/virologia , Macrófagos Alveolares , Peste dos Pequenos Ruminantes/imunologia , Peste dos Pequenos Ruminantes/metabolismo , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/crescimento & desenvolvimento , Vírus da Peste dos Pequenos Ruminantes/imunologia , Vírus da Peste dos Pequenos Ruminantes/metabolismo , Ligação Proteica , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Proteínas Virais de Fusão/metabolismo
2.
Proteins ; 92(3): 356-369, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37881117

RESUMO

The fusion of haemagglutinin-neuraminidase (HN) protein of peste des petits ruminant (PPR) virus with signaling lymphocyte activation molecules (SLAM) host cell receptor consequences the virus entry and multiplication inside the host cell. The use of synthetic SLAM homologous peptides (i.e., molecular decoy for HN protein of PPR virus) may check PPR infection at the preliminary stage. Hence, the predicted SLAM homologous peptides using bioinformatics tools were synthesized by solid phase chemistry with standard Merrifield's 9-fluorenylmethoxycarbonyl (Fmoc) chemistry and were purified by reverse phase high performance liquid chromatography. The secondary structures of synthesized peptides were elucidated by circular dichroism spectroscopy. The in vitro interactions of these peptides were studied through indirect Enzyme Linked Immuno Sorbent Assay (ELISA) and visual surface plasmon UV-visible spectroscopy. The SLAM homologous peptides were able to interact with the peste des petits ruminant virus (PPRV) with varying binding efficiency. The interaction of SLAM homologous peptide with the PPR virus was ascertained by the change in the plasmon color from red wine to purple during visual detection and also by bathochromic shift in absorbance spectra under UV-visible spectrophotometry. The cytotoxic and anti-PPRV effect of these peptides were also evaluated in B95a cell line using PPR virus (Sungri/96). The cytotoxic concentration 50 (CC50 ) value of each peptide was greater than 1000 µg mL-1 . The anti-PPRV efficiency of SLAM-22 was relatively high among SLAM homologous peptides, SLAM-22 at 25 µg mL-1 concentration showed a reduction of more than log10 3 virus titer by priming of B95a cell line while the use of SLAM-15 and Muco-17 at the same concentration dropped virus titer from log10 4.8 to log10 2.5 and log10 3.1 respectively. The concentration of SLAM homologous peptide (25 µg mL-1 ) to exert its anti-PPRV effect was much less than its CC50 level (>1000 µg mL-1 ). Therefore, the synthetic SLAM homologous peptides may prove to be better agents to target PPRV.


Assuntos
Peste dos Pequenos Ruminantes , Vírus da Peste dos Pequenos Ruminantes , Animais , Vírus da Peste dos Pequenos Ruminantes/metabolismo , Peste dos Pequenos Ruminantes/metabolismo , Linhagem Celular , Proteínas Virais/metabolismo , Peptídeos/farmacologia , Peptídeos/metabolismo , Cabras
3.
J Gen Virol ; 104(10)2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37831061

RESUMO

Peste des petits ruminants virus (PPRV) is known to induce transient immunosuppression in infected small ruminants by modulating several cellular pathways involved in the antiviral immune response. Our study shows that the PPRV-coded non-structural proteins C and V can interact with the cellular NF-κB p65 subunit. The PPRV-C protein interacts with the transactivation domain (TAD) while PPRV-V interacts with the Rel homology domain (RHD) of the NF-κB p65 subunit. Both viral proteins can suppress the NF-κB transcriptional activity and NF-κB-mediated transcription of cellular genes. PPRV-V protein expression can significantly inhibit the nuclear translocation of NF-κB p65 upon TNF-α stimulation, whereas PPRV-C does not affect it. The NF-κB-mediated pro-inflammatory cytokine gene expression is significantly downregulated in cells expressing PPRV-C or PPRV-V protein. Our study provides evidence suggesting a role of PPRV non-structural proteins V and C in the modulation of NF-κB signalling through interaction with the NF-κB p65 subunit.


Assuntos
Peste dos Pequenos Ruminantes , Vírus da Peste dos Pequenos Ruminantes , Animais , Humanos , Vírus da Peste dos Pequenos Ruminantes/genética , Peste dos Pequenos Ruminantes/metabolismo , Citocinas/genética , Citocinas/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Ruminantes , Expressão Gênica , Cabras/genética
4.
J Gen Virol ; 101(1): 33-43, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31794379

RESUMO

Peste des petits ruminants (PPR) is a highly contagious disease of small ruminants that is caused by peste des petits ruminants virus (PPRV). To date, the molecular mechanism of PPRV infection is still unclear. It is well known that host proteins might be involved in the pathogenesis process for many viruses. In this study, we first proved that nucleolin (NCL), a highly conserved host factor, interacts with the core domain of PPRV N protein through its C terminus and co-locates with the N protein in the nucleus of cells. To investigate the role of NCL in PPRV infection, the expression level of NCL was inhibited with small interfering RNAs of NCL, and the results showed that PPRV growth was improved. However, the proliferation of PPRV was inhibited when the expression level of NCL was improved. Further analysis indicated that the inhibitory effect of NCL on the PPRV was caused by stimulating the interferon (IFN) pathways in host cells. In summary, our results will help us to understand the mechanism of PPRV infection.


Assuntos
Peste dos Pequenos Ruminantes/metabolismo , Vírus da Peste dos Pequenos Ruminantes/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ruminantes/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Células HEK293 , Humanos , Interferons/metabolismo , Proteínas do Nucleocapsídeo/metabolismo , Ruminantes/virologia , Células Vero , Nucleolina
5.
J Gen Virol ; 101(3): 242-251, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31859612

RESUMO

Peste-des-petits-ruminants virus (PPRV) haemagglutinin (H) protein mediates binding to cellular receptors and then initiates virus entry. To identify the key residues of PPRV H (Hv) protein of the Nigeria 75/1 strain involved in binding to receptors, interaction of the Hv and mutated Hv (mHv) proteins with receptors (SLAM and Nectin 4) and their mutants (mSLAM1, mSLAM2, mSLAM3 and mNectin 4) was investigated using surface plasmon resonance imaging (SPRi) and coimmunoprecipitation (co-IP) assays. The results showed that the Hv protein failed to interact with mSLAM3, but interacted at a strong or medium intensity with SLAM, mSLAM2, Nectin 4 and mNectin 4, and at a low level with mSLAM1. The mHv protein was unable to interact with SLAM and its mutants, but bound to Nectin 4 and mNectin 4 with medium and weak intensity, respectively. Further analysis showed that the Hv protein could precipitate mSLAM1, mSLAM2 and mNectin 4, but not mSLAM3. The mHv protein failed to coprecipitate with SLAM and its mutants. The binding activities of mNectin 4 and Nectin 4 to mHv were less than 30.36 and 51.94 % of the wild-type levels, respectively. Based on the results obtained, amino acids at positions R389, L464, I498, R503, R533, Y541, Y543, F552 and Y553 of H protein and I61, H62, L64, K76, K78, E123, H130, I210, A211, S226 and R227 in SLAM were identified to be essential for the specificity of H-SLAM interaction, while the critical residues of H-Nectin 4 interaction require further study. These findings would improve our understanding of the invasive mechanisms of PPRV.


Assuntos
Aminoácidos/análise , Moléculas de Adesão Celular/metabolismo , Hemaglutininas Virais/metabolismo , Peste dos Pequenos Ruminantes/metabolismo , Vírus da Peste dos Pequenos Ruminantes/metabolismo , Receptores de Superfície Celular/metabolismo , Proteína Associada à Molécula de Sinalização da Ativação Linfocitária/metabolismo , Sequência de Aminoácidos , Aminoácidos/química , Aminoácidos/metabolismo , Animais , Células CHO , Moléculas de Adesão Celular/genética , Cricetulus , Células HEK293 , Humanos , Proteínas Mutantes/metabolismo , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/química , Receptores de Superfície Celular/genética , Proteínas Recombinantes/metabolismo , Proteína Associada à Molécula de Sinalização da Ativação Linfocitária/genética , Transfecção , Internalização do Vírus
6.
J Virol ; 93(16)2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31167907

RESUMO

Peste des petits ruminants virus (PPRV) is the etiological agent of peste des petits ruminants, causing acute immunosuppression in its natural hosts. However, the molecular mechanisms by which PPRV antagonizes the host immune responses have not been fully characterized. In particular, how PPRV suppresses the activation of the host RIG-I-like receptor (RLR) pathway has yet to be clarified. In this study, we demonstrated that PPRV infection significantly suppresses RLR pathway activation and type I interferon (IFN) production and identified PPRV N protein as an extremely important antagonistic viral factor that suppresses beta interferon (IFN-ß) and IFN-stimulated gene (ISG) expression. A detailed analysis showed that PPRV N protein inhibited type I IFN production by targeting interferon regulatory factor 3 (IRF3), a key molecule in the RLR pathway required for type I IFN induction. PPRV N protein interacted with IRF3 (but not with other components of the RLR pathway, including MDA5, RIG-I, VISA, TBK1, and MITA) and abrogated the phosphorylation of IRF3. As expected, PPRV N protein also considerably impaired the nuclear translocation of IRF3. The TBK1-IRF3 interaction was involved significantly in IRF3 phosphorylation, and we showed that PPRV N protein inhibits the association between TBK1 and IRF3, which in turn inhibits IRF3 phosphorylation. The amino acid region 106 to 210 of PPRV N protein was determined to be essential for suppressing the nuclear translocation of IRF3 and IFN-ß production, and the 140 to 400 region of IRF3 was identified as the crucial region for the N-IRF3 interaction. Together, our findings demonstrate a new mechanism evolved by PPRV to inhibit type I IFN production and provide structural insights into the immunosuppression caused by PPRV.IMPORTANCE Peste des petits ruminants is a highly contagious animal disease affecting small ruminants, which threatens both small livestock and endangered susceptible wildlife populations in many countries. The causative agent, peste des petits ruminants virus (PPRV), often causes acute immunosuppression in its natural hosts during infection. Here, for the first time, we demonstrate that N protein, the most abundant protein of PPRV, plays an extremely important role in suppression of interferon regulatory factor 3 (IRF3) function and type I interferon (IFN) production by interfering with the formation of the TBK1-IRF3 complex. This study explored a novel antagonistic mechanism of PPRV.


Assuntos
Interações Hospedeiro-Patógeno , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/biossíntese , Proteínas do Nucleocapsídeo/metabolismo , Peste dos Pequenos Ruminantes/metabolismo , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Animais , Imunomodulação , Interferon beta/genética , Fosforilação , Regiões Promotoras Genéticas , Ligação Proteica , Transporte Proteico , Ativação Transcricional
7.
Virology ; 595: 110056, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38552409

RESUMO

The Peste des petits ruminant virus (PPRV) is a member of the Paramyxoviridae family and is classified into the genus Measles virus. PPRV predominantly infects small ruminants, leading to mortality rates of nearly 100%, which have caused significant economic losses in developing countries. Host proteins are important in virus replication, but the PPRV nucleocapsid (N) protein-host interacting partners for regulating PPRV replication remain unclear. The present study confirmed the interaction between PPRV-N and the host protein vimentin by co-immunoprecipitation and co-localization experiments. Overexpression of vimentin suppressed PPRV replication, whereas vimentin knockdown had the opposite effect. Mechanistically, N was subjected to degradation via the ubiquitin/proteasome pathway, where vimentin recruits the E3 ubiquitin ligase NEDD4L to fulfill N-ubiquitination, resulting in the degradation of the N protein. These findings suggest that the host protein vimentin and E3 ubiquitin ligase NEDD4L have an anti-PPRV effect.


Assuntos
Proteínas do Nucleocapsídeo , Vírus da Peste dos Pequenos Ruminantes , Vimentina , Replicação Viral , Proteínas do Nucleocapsídeo/metabolismo , Proteínas do Nucleocapsídeo/genética , Vimentina/metabolismo , Vimentina/genética , Animais , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Vírus da Peste dos Pequenos Ruminantes/genética , Vírus da Peste dos Pequenos Ruminantes/metabolismo , Humanos , Ubiquitinação , Interações Hospedeiro-Patógeno , Células HEK293 , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Ubiquitina-Proteína Ligases Nedd4/genética , Linhagem Celular , Peste dos Pequenos Ruminantes/virologia , Peste dos Pequenos Ruminantes/metabolismo , Ligação Proteica
8.
Microbiol Spectr ; 10(5): e0103122, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36036587

RESUMO

Peste des petits ruminants virus (PPRV) causes a highly contagious disease in small ruminants and severe economic losses in developing countries. PPRV infection can stimulate high levels of interferon (IFN) and many IFN-stimulated genes (ISGs), such as ISG15, which may play a key role in the process of viral infection. However, the role of ISG15 in PPRV infection and replication has not yet been reported. In this study, we found ISG15 expression to be significantly upregulated after PPRV infection of caprine endometrial epithelial cells (EECs), and ISG15 inhibits the proliferation of PPRV. Further analysis showed that free ISG15 could inhibit PPRV proliferation. Moreover, ISG15 does not affect the binding, entry, and transcription but does suppress the replication of PPRV. A detailed analysis revealed that ISG15 interacts and colocalizes with both viral N and P proteins and that its interactive regions are all located in the N-terminal domain. Further studies showed that ISG15 can competitively interact with N and P proteins and significantly interfere with their binding. Finally, through the construction of the C-terminal mutants of ISG15 with different lengths, it was found that amino acids (aa) 77 to 101 play a key role in inhibiting the binding of N and P proteins and that interaction with the P protein disappears after the deletion of 77 to 101 aa. The present study revealed a novel mechanism of ISG15 in disrupting the activity of the N0-P complex to inhibit viral replication. IMPORTANCE PPRV, a widespread and fatal disease of small ruminants, is one of the most devastating animal diseases in Africa, the Middle East, and Asia, causing severe economic losses. IFNs play an important role as a component of natural immunity against pathogens, yet the role of ISG15, an IFN-stimulated gene, in protecting against PPRV infection is currently unknown. We demonstrated, for the first time, that free ISG15 inhibits PPRV proliferation by disrupting the activity of the N0-P complex, a finding that has not been reported in other viruses. Our results provide important insights that can further understand the pathogenesis and innate immune mechanisms of PPRV.


Assuntos
Peste dos Pequenos Ruminantes , Vírus da Peste dos Pequenos Ruminantes , Animais , Vírus da Peste dos Pequenos Ruminantes/genética , Peste dos Pequenos Ruminantes/metabolismo , Nucleoproteínas , Fosfoproteínas , Cabras , Interferons/genética , Ruminantes , Aminoácidos
9.
Autophagy ; 16(5): 842-861, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31318632

RESUMO

Macroautophagy/autophagy is an essential cellular response in the fight against intracellular pathogens. Although some viruses can escape from or utilize autophagy to ensure their own replication, the responses of autophagy pathways to viral invasion remain poorly documented. Here, we show that peste des petits ruminants virus (PPRV) infection induces successive autophagic signalling in host cells via distinct and uncoupled molecular pathways. Immediately upon invasion, PPRV induced a first transient wave of autophagy via a mechanism involving the cellular pathogen receptor NECTIN4 and an AKT-MTOR-dependent pathway. Autophagic detection showed that early PPRV infection not only increased the amounts of autophagosomes and LC3-II but also downregulated the phosphorylation of AKT-MTOR. Subsequently, we found that the binding of viral protein H to NECTIN4 ultimately induced a wave of autophagy and inactivated the AKT-MTOR pathway, which is a critical step for the control of infection. Soon after infection, new autophagic signalling was initiated that required viral replication and protein expression. Interestingly, expression of IRGM and HSPA1A was significantly upregulated following PPRV replication. Strikingly, knockdown of IRGM and HSPA1A expression using small interfering RNAs impaired the PPRV-induced second autophagic wave and viral particle production. Moreover, IRGM-interacting PPRV-C and HSPA1A-interacting PPRV-N expression was sufficient to induce autophagy through an IRGM-HSPA1A-dependent pathway. Importantly, syncytia formation could facilitate sustained autophagy and the replication of PPRV. Overall, our work reveals distinct molecular pathways underlying the induction of self-beneficial sustained autophagy by attenuated PPRV, which will contribute to improving the use of vaccines for therapy.Abbreviations: ACTB: actin beta; ANOVA: analysis of variance; ATG: autophagy-related; BECN1: beclin 1; CDV: canine distemper virus; Co-IP: coimmunoprecipitation; FIP: fusion inhibitory peptide; GFP: green fluorescent protein; GST: glutathione S-transferase; HMOX1: heme oxygenase 1; hpi: hours post infection; HSPA1A: heat shock protein family A (Hsp70) member 1A; HSP90AA1: heat shock protein 90 kDa alpha (cytosolic), class A member 1; IFN: interferon; IgG: immunoglobulin G; INS: insulin; IRGM: immunity related GTPase M; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MeV: measles virus; MOI: multiplicity of infection; MTOR: mechanistic target of rapamycin kinase; PI3K: phosphoinositide-3 kinase; PIK3C3: phosphatidylinositol 3-kinase catalytic subunit type 3; SDS: sodium dodecyl sulfate; siRNA: small interfering RNA; SQSTM1/p62: sequestosome 1; UV: ultraviolet.


Assuntos
Autofagossomos/metabolismo , Autofagia/fisiologia , Lisossomos/metabolismo , Peste dos Pequenos Ruminantes/metabolismo , Animais , Interferons/metabolismo , Peste dos Pequenos Ruminantes/imunologia , Vírus da Peste dos Pequenos Ruminantes/genética , Vírus da Peste dos Pequenos Ruminantes/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/fisiologia , Replicação Viral/fisiologia
10.
Front Immunol ; 10: 3066, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32038620

RESUMO

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 Viral
11.
Viruses ; 11(8)2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31398809

RESUMO

Peste des Petits Ruminant (PPR) is an important transboundary, OIE-listed contagious viral disease of primarily sheep and goats caused by the PPR virus (PPRV), which belongs to the genus Morbillivirus of the family Paramyxoviridae. The mortality rate is 90-100%, and the morbidity rate may reach up to 100%. PPR is considered economically important as it decreases the production and productivity of livestock. In many endemic poor countries, it has remained an obstacle to the development of sustainable agriculture. Hence, proper control measures have become a necessity to prevent its rapid spread across the world. For this, detailed information on the pathogenesis of the virus and the virus host interaction through cellular receptors needs to be understood clearly. Presently, two cellular receptors; signaling lymphocyte activation molecule (SLAM) and Nectin-4 are known for PPRV. However, extensive information on virus interactions with these receptors and their impact on host immune response is still required. Hence, a thorough understanding of PPRV receptors and the mechanism involved in the induction of immunosuppression is crucial for controlling PPR. In this review, we discuss PPRV cellular receptors, viral host interaction with cellular receptors, and immunosuppression induced by the virus with reference to other Morbilliviruses.


Assuntos
Interações Hospedeiro-Patógeno , Peste dos Pequenos Ruminantes/metabolismo , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Receptores de Superfície Celular/metabolismo , Receptores Virais/metabolismo , Animais , Genoma Viral , Genômica/métodos , Especificidade de Hospedeiro , Interações Hospedeiro-Patógeno/imunologia , Imunidade , Peste dos Pequenos Ruminantes/imunologia , Ligação Proteica , Receptores de Superfície Celular/química , Receptores Virais/química , Transdução de Sinais , Relação Estrutura-Atividade , Proteínas Virais/química , Proteínas Virais/metabolismo
12.
PLoS One ; 12(5): e0177300, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28475628

RESUMO

Peste des petits ruminants virus (PPRV) is a morbillivirus that produces clinical disease in goats and sheep. We have studied the induction of interferon-ß (IFN-ß) following infection of cultured cells with wild-type and vaccine strains of PPRV, and the effects of such infection with PPRV on the induction of IFN-ß through both MDA-5 and RIG-I mediated pathways. Using both reporter assays and direct measurement of IFN-ß mRNA, we have found that PPRV infection induces IFN-ß only weakly and transiently, and the virus can actively block the induction of IFN-ß. We have also generated mutant PPRV that lack expression of either of the viral accessory proteins (V&C) to characterize the role of these proteins in IFN-ß induction during virus infection. Both PPRV_ΔV and PPRV_ΔC were defective in growth in cell culture, although in different ways. While the PPRV V protein bound to MDA-5 and, to a lesser extent, RIG-I, and over-expression of the V protein inhibited both IFN-ß induction pathways, PPRV lacking V protein expression can still block IFN-ß induction. In contrast, PPRV C bound to neither MDA-5 nor RIG-I, but PPRV lacking C protein expression lost the ability to block both MDA-5 and RIG-I mediated activation of IFN-ß. These results shed new light on the inhibition of the induction of IFN-ß by PPRV.


Assuntos
Fibroblastos/metabolismo , Doenças das Cabras/virologia , Interferon Tipo I/metabolismo , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/metabolismo , Animais , Fibroblastos/virologia , Doenças das Cabras/metabolismo , Cabras , Peste dos Pequenos Ruminantes/metabolismo
13.
J Comp Pathol ; 131(2-3): 135-41, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15276853

RESUMO

This report describes the pathological and immunohistochemical findings in naturally infected lambs from three outbreaks of peste des petits ruminants in Mugla and Aydin provinces of the Ege district of Turkey. At necropsy, ulcerative stomatitis, catarrhal or fibrinous bronchopneumonia, and acute catarrhal enteritis were observed. Histopathologically, syncytial cells containing inclusion bodies were seen in the tongue and in the buccal, labial and soft palate mucosae. In pneumonic lungs, syncytial cells were present in the alveolar lumina, and cytoplasmic inclusion bodies in epithelial cells of the bronchi and bronchioli. Immunohistochemically, viral antigen was strongly labelled in the lung, oral tissues and small intestine.


Assuntos
Peste dos Pequenos Ruminantes/patologia , Peste dos Pequenos Ruminantes/veterinária , Animais , Antígenos Virais/imunologia , Antígenos Virais/metabolismo , Feminino , Imuno-Histoquímica , Masculino , Peste dos Pequenos Ruminantes/metabolismo , Vírus da Peste dos Pequenos Ruminantes , Ovinos , Turquia
14.
Virus Res ; 145(1): 9-17, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19540279

RESUMO

The large (L) polymerase gene and the 5'-terminal UTR of the genome of peste des petits ruminants virus (PPRV), vaccine strain Nigeria 75/1, were cloned and sequenced. The L protein was also expressed in eukaryotic cells and its polymerase activity was quantitatively measured in a PPR reverse genetics assay using a reporter minigenome. Comparative sequence analysis of this functional L gene with corresponding genes of other morbilliviruses showed a degree of conservation exceeding 70%. The multiple sequence alignment and the phylogenetic study of L gene discriminated the morbilliviruses in 6 clusters, which are more closely related to Tupaia and Henipaviruses than to other paramyxoviruses. Important protein domains and functional motifs of the L polymerase of the PPRV Nigeria 75/1 vaccine were also identified by using different bioinformatics tools.


Assuntos
Genes Virais , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/genética , Proteínas Virais/genética , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , RNA Polimerases Dirigidas por DNA/biossíntese , RNA Polimerases Dirigidas por DNA/genética , Genoma Viral , Dados de Sequência Molecular , Peste dos Pequenos Ruminantes/metabolismo , Vírus da Peste dos Pequenos Ruminantes/metabolismo , Filogenia , RNA Viral/análise , RNA Viral/genética , Alinhamento de Sequência , Análise de Sequência de RNA , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA