Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Virology ; 595: 110056, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38552409

RESUMEN

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.


Asunto(s)
Proteínas de la Nucleocápside , Virus de la Peste de los Pequeños Rumiantes , Vimentina , Replicación Viral , Proteínas de la Nucleocápside/metabolismo , Proteínas de la Nucleocápside/genética , Vimentina/metabolismo , Vimentina/genética , Animales , Virus de la Peste de los Pequeños Rumiantes/fisiología , Virus de la Peste de los Pequeños Rumiantes/genética , Virus de la Peste de los Pequeños Rumiantes/metabolismo , Humanos , Ubiquitinación , Interacciones Huésped-Patógeno , Células HEK293 , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Ubiquitina-Proteína Ligasas Nedd4/genética , Línea Celular , Peste de los Pequeños Rumiantes/virología , Peste de los Pequeños Rumiantes/metabolismo , Unión Proteica
2.
J Virol ; 97(2): e0171222, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36651745

RESUMEN

The pathogenic mechanisms of peste des petits ruminants virus (PPRV) infection remain poorly understood, leaving peste des petits ruminants (PPR) control and eradication especially difficult. Here, we determined that PPRV nucleocapsid (N) protein triggers formation of stress granules (SGs) to benefit viral replication. A mass spectrometry-based profiling of the interactome of PPRV N protein revealed that PPRV N protein interacted with protein kinase R (PKR)-activating protein (PACT), and this interaction was confirmed in the context of PPRV infection. PACT was essential for PPRV replication. Besides, the ectopic expression of N activated the PKR/eIF2α (α subunit of eukaryotic initiation factor 2) pathway through induction of PKR phosphorylation, but it did not induce PKR phosphorylation in PACT-deficient (PACT-/-) cells. PPRV N interacted with PACT, impairing the interaction between PACT and a PKR inhibitor, transactivation response RNA-binding protein (TRBP), which subsequently enhanced the interaction between PACT and PKR and thus promoted the activation of PKR and eIF2α phosphorylation, resulting in formation of stress granules (SGs). Consistently, PPRV infection induced SG formation through activation of the PKR/eIF2α pathway, and knockdown of N impaired PPRV-induced SG formation. PPRV-induced SG formation significantly decreased in PACT-/- cells as well. The role of SG formation in PPRV replication was subsequently investigated, which showed that SG formation plays a positive role in PPRV replication. By using an RNA fluorescence in situ hybridization assay, we found that PPRV-induced SGs hid cellular mRNA rather than viral mRNA. Altogether, our data provide the first evidence that PPRV N protein plays a role in modulating the PKR/eIF2α/SG axis and promotes virus replication through targeting PACT. IMPORTANCE Stress granule (SG) formation is a conserved cellular strategy to reduce stress-related damage regulating cell survival. A mass spectrometry-based profiling of the interactome of PPRV N protein revealed that PPRV N interacted with PACT to regulate the assembly of SGs. N protein inhibited the interaction between PACT and a PKR inhibitor, TRBP, through binding to the M1 domain of PACT, which enhanced the interaction between PACT and PKR and thus promoted PKR activation and subsequent eIF2α phosphorylation as well as SG formation. The regulatory function of N protein was strikingly abrogated in PACT-/- cells. SGs induced by PPRV infection through the PKR/eIF2α pathway are PACT dependent. The loss-of-function assay indicated that PPRV-induced SGs were critical for PPRV replication. We concluded that the PPRV N protein manipulates the host PKR/eIF2α/SG axis to favor virus replication.


Asunto(s)
Proteínas de la Nucleocápside , Peste de los Pequeños Rumiantes , Virus de la Peste de los Pequeños Rumiantes , Proteínas de Unión al ARN , Gránulos de Estrés , Replicación Viral , Animales , Humanos , Hibridación Fluorescente in Situ , Proteínas de la Nucleocápside/metabolismo , Peste de los Pequeños Rumiantes/fisiopatología , Virus de la Peste de los Pequeños Rumiantes/fisiología , Proteínas Quinasas/metabolismo , Proteínas de Unión al ARN/metabolismo , Gránulos de Estrés/metabolismo , Replicación Viral/genética
3.
Vet Res ; 53(1): 89, 2022 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-36307867

RESUMEN

Peste des petits ruminants (PPR) is an acute and highly contagious disease and has long been a significant threat to small ruminant productivity worldwide. However, the molecular mechanism underlying host-PPRV interactions remains unclear and the long noncoding RNAs (lncRNAs) regulation of PPR virus (PPRV) infection has rarely been reported so far. Here, we first demonstrated that PPRV infection can induce an obvious innate immune response in caprine endometrial epithelial cells (EECs) at 48 h post-infection (hpi) with an MOI of 3. Subsequently, we determined that PPRV infection is associated with 191 significantly differentially expressed (SDE) lncRNAs, namely, 137 upregulated and 54 downregulated lncRNAs, in caprine EECs compared with mock control cells at 48 hpi by using deep sequencing technology. Importantly, bioinformatics preliminarily analyses revealed that these DE lncRNAs were closely related to the immune response. Furthermore, we identified a system of lncRNAs related to the immune response and focused on the role of lncRNA 10636385 (IRF1-AS) in regulating the innate immune response. Interestingly, we found that IRF1-AS was a potent positive regulator of IFN-ß and ISG production, which can significantly inhibit PPRV replication in host cells. In addition, our data revealed that IRF1-AS was positively correlated with its potential target gene, IRF1, which enhanced the activation of IRF3 and the expression of ISGs and interacted with IRF3. This study suggests that IRF1-AS could be a new host factor target for developing antiviral therapies against PPRV infection.


Asunto(s)
Enfermedades de las Cabras , Peste de los Pequeños Rumiantes , Virus de la Peste de los Pequeños Rumiantes , ARN Largo no Codificante , Animales , Peste de los Pequeños Rumiantes/genética , ARN Largo no Codificante/genética , Cabras/genética , Virus de la Peste de los Pequeños Rumiantes/fisiología , Interferón beta
4.
Vet Microbiol ; 270: 109451, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35594636

RESUMEN

Peste des petits ruminants virus (PPRV) has long been a significant threat to small ruminant productivity worldwide. Virus infection-induced endoplasmic reticulum (ER) stress (ERS) and the subsequently activated unfolded protein response (UPR) play significant roles in viral replication and pathogenesis. However, the relationship between ERS and PPRV infection is unknown. In this study, we demonstrated that ERS was induced during PPRV infection in caprine endometrial epithelial cells (EECs). Importantly, we demonstrated that the induction of autophagy by PPRV was mediated by ERS. Furthermore, we found that the PERK/eIF2α pathway but not the ATF6 or IRE1 pathway was activated and that the activated PERK/eIF2α pathway participated in regulating ERS-mediated autophagy. Moreover, virus replication was required for PPRV infection-induced ERS-mediated autophagy and PERK pathway activation. Additionally, we revealed that either the viral nucleocapsid (N) or nonstructural protein C was sufficient to elicit ERS and activate the PERK/eIF2α pathway, which further increased autophagy. Taken together, these results suggest that PPRV N and C protein-induced autophagy enhances viral replication through the induction of ERS and that the PERK pathway may be involved in the activation of ERS-mediated autophagy during PPRV infection.


Asunto(s)
Enfermedades de las Cabras , Peste de los Pequeños Rumiantes , Virus de la Peste de los Pequeños Rumiantes , Animales , Autofagia , Virus ADN , Factor 2 Eucariótico de Iniciación , Cabras , Virus de la Peste de los Pequeños Rumiantes/fisiología , Rumiantes , Replicación Viral/fisiología
5.
Vet Microbiol ; 260: 109186, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34333402

RESUMEN

Replication of peste des petits ruminants virus (PPRV) strongly depends on the cellular environment and resources of host cells including nucleoside pool. Thus, enzymes involved in nucleoside biosynthesis (such as pyrimidine biosynthesis pathway) are regarded as attractive targets for antiviral drug development. Here, we demonstrate that brequinar (BQR) and leflunomide (LFM) which are two specific inhibitors of DHODH enzyme and 6-azauracil (6-AU) which is an ODase enzyme inhibitor robustly inhibit PPRV replication in HEK293T cell line as well as in peripheral blood mononuclear cells isolated from goat. We further demonstrate that these agents exert anti-PPRV activity via the depletion of purimidine nucleotide. Interestingly, these inhibitors can trigger the transcription of antiviral interferon-stimulated genes (ISGs). However, the induction of ISGs is largely independent of the classical JAK-STAT pathway. Combination of BQR with interferons (IFNs) exerts enhanced ISG induction and anti-PPRV activity. Taken together, this study reveals an unconventional novel mechanism of crosstalk between nucleotide biosynthesis pathways and cellular antiviral immunity in inhibiting PPRV replication. In conclusion, targeting pyrimidine biosynthesis represents a potential strategy for developing antiviral strategies against PPRV.


Asunto(s)
Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , Nucleósidos/metabolismo , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes/fisiología , Animales , Compuestos de Bifenilo/farmacología , Células HEK293 , Humanos , Inmunidad Celular , Interferones/farmacología , Leflunamida/farmacología , Leucocitos Mononucleares/inmunología , Virus de la Peste de los Pequeños Rumiantes/efectos de los fármacos , Virus de la Peste de los Pequeños Rumiantes/inmunología , Pirimidinas/metabolismo , Uracilo/análogos & derivados , Uracilo/farmacología , Replicación Viral
6.
Viruses ; 13(5)2021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-34066336

RESUMEN

Peste des petits ruminants (PPR) is a viral disease of goats and sheep that occurs in Africa, the Middle East and Asia with a severe impact on livelihoods and livestock trade. Many wild artiodactyls are susceptible to PPR virus (PPRV) infection, and some outbreaks have threatened endangered wild populations. The role of wild species in PPRV epidemiology is unclear, which is a knowledge gap for the Global Strategy for the Control and Eradication of PPR. These studies aimed to investigate PPRV infection in wild artiodactyls in the Greater Serengeti and Amboseli ecosystems of Kenya and Tanzania. Out of 132 animals purposively sampled in 2015-2016, 19.7% were PPRV seropositive by ID Screen PPR competition enzyme-linked immunosorbent assay (cELISA; IDvet, France) from the following species: African buffalo, wildebeest, topi, kongoni, Grant's gazelle, impala, Thomson's gazelle, warthog and gerenuk, while waterbuck and lesser kudu were seronegative. In 2018-2019, a cross-sectional survey of randomly selected African buffalo and Grant's gazelle herds was conducted. The weighted estimate of PPRV seroprevalence was 12.0% out of 191 African buffalo and 1.1% out of 139 Grant's gazelles. All ocular and nasal swabs and faeces were negative by PPRV real-time reverse transcription-polymerase chain reaction (RT-qPCR). Investigations of a PPR-like disease in sheep and goats confirmed PPRV circulation in the area by rapid detection test and/or RT-qPCR. These results demonstrated serological evidence of PPRV infection in wild artiodactyl species at the wildlife-livestock interface in this ecosystem where PPRV is endemic in domestic small ruminants. Exposure to PPRV could be via spillover from infected small ruminants or from transmission between wild animals, while the relatively low seroprevalence suggests that sustained transmission is unlikely. Further studies of other major wild artiodactyls in this ecosystem are required, such as impala, Thomson's gazelle and wildebeest.


Asunto(s)
Animales Salvajes/virología , Ecosistema , Ganado/virología , Peste de los Pequeños Rumiantes/epidemiología , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes/fisiología , Enfermedades de los Animales/epidemiología , Enfermedades de los Animales/historia , Enfermedades de los Animales/virología , Animales , Estudios Transversales , Brotes de Enfermedades , Geografía Médica , Historia del Siglo XXI , Kenia/epidemiología , Peste de los Pequeños Rumiantes/historia , Virus de la Peste de los Pequeños Rumiantes/clasificación , Vigilancia en Salud Pública , Estudios Seroepidemiológicos , Tanzanía/epidemiología
7.
J Immunol ; 206(3): 566-579, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33380495

RESUMEN

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.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Fibroblastos/fisiología , Factor 3 Regulador del Interferón/metabolismo , Peste de los Pequeños Rumiantes/inmunología , Virus de la Peste de los Pequeños Rumiantes/fisiología , Fosfoproteínas/metabolismo , Proteínas Virales/metabolismo , Animales , Células Cultivadas , Cabras , Humanos , Evasión Inmune , Inmunidad Innata , Factor 3 Regulador del Interferón/genética , Transducción de Señal , Replicación Viral
8.
Vet Med Sci ; 7(2): 393-401, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33010117

RESUMEN

BACKGROUND: Knowledge of sequential changes in haematobiochemical parameters of infected animals helps in the formulation of appropriate supportive therapy. OBJECTIVE: We investigated the sequential haematological and biochemical changes in peste des petits ruminants (PPR)-infected Black Bengal goats. METHODS: Goats were either infected with PPR virus (PPRV; n = 8) or sham infected with sterile phosphate-buffered saline (n = 4) via the intranasal route. Blood and sera were collected from both groups at different days post-infection (dpi) and analysed. Goats were sacrificed at different dpi and the amount of PPRV RNA in different tissues was quantified by real-time RT-PCR. RESULTS: The PPRV-infected goats showed mild depression and scanty nasal secretions starting at 4 dpi which became severe with high fever (106°F), dyspnoea, stomatitis, profuse orinasal discharge and diarrhoea at 9-13 dpi. PPRV RNA was detected in different tissues of infected goats. Severe lymphocytic leukopenia (at 18 dpi) was observed in infected goats. Total protein and albumin decreased in infected goats starting at 10 dpi. An elevated level of enzymes (alkaline phosphatase, creatine kinase, aspartate transaminase and alanine transaminase) and metabolites (blood urea nitrogen and urea B) were found in infected goats starting at 7-10 dpi, suggesting damages in the liver and kidneys. PPR-infected goats showed elevated sodium and chloride ions starting at 7 dpi. The majority of infected goats were seroconverted by 14 dpi. CONCLUSIONS: Anti-diarrheal agents, aqua solutions and other medicine to support liver and kidney functions could be considered as supportive therapy against PPRV infection.


Asunto(s)
Enfermedades de las Cabras/sangre , Peste de los Pequeños Rumiantes/sangre , Animales , Bangladesh , Enfermedades de las Cabras/virología , Cabras , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes/fisiología
9.
Viruses ; 12(3)2020 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-32156067

RESUMEN

In the recent past, peste des petits ruminants (PPR) emerged in East Africa causing outbreaks in small livestock across different countries, with evidences of spillover to wildlife. In order to understand better PPR at the wildlife-livestock interface, we investigated patterns of peste des petits ruminants virus (PPRV) exposure, disease outbreaks, and viral sequences in the northern Albertine Rift. PPRV antibodies indicated a widespread exposure in apparently healthy wildlife from South Sudan (2013) and Uganda (2015, 2017). African buffaloes and Uganda kobs <1-year-old from Queen Elizabeth National Park (2015) had antibodies against PPRV N-antigen and local serosurvey captured a subsequent spread of PPRV in livestock. Outbreaks with PPR-like syndrome in sheep and goats were recorded around the Greater Virunga Landscape in Kasese (2016), Kisoro and Kabale (2017) from western Uganda, and in North Kivu (2017) from eastern Democratic Republic of the Congo (DRC). This landscape would not be considered typical for PPR persistence as it is a mixed forest-savannah ecosystem with mostly sedentary livestock. PPRV sequences from DRC (2017) were identical to strains from Burundi (2018) and confirmed a transboundary spread of PPRV. Our results indicate an epidemiological linkage between epizootic cycles in livestock and exposure in wildlife, denoting the importance of PPR surveillance on wild artiodactyls for both conservation and eradication programs.


Asunto(s)
Animales Salvajes/virología , Ganado/virología , Peste de los Pequeños Rumiantes/epidemiología , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes , África Oriental/epidemiología , Animales , Anticuerpos Antivirales/inmunología , Brotes de Enfermedades , Ensayo de Inmunoadsorción Enzimática , Femenino , Geografía Médica , Cabras , Masculino , Virus de la Peste de los Pequeños Rumiantes/clasificación , Virus de la Peste de los Pequeños Rumiantes/fisiología , Estudios Seroepidemiológicos , Ovinos
10.
Microb Pathog ; 140: 103949, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31875517

RESUMEN

Peste des petits ruminant (PPR), a highly contagious viral disease of small ruminants, is characterized by erosive stomatitis and pneumo-enteritis. However, its neurovirulence potential as observed with other morbilliviruses has not been fully investigated. The present study describes the neuropathological alterations induced by PPR virus through apoptotic pathway. A total number of 12 carcasses of local breed goat kids of either sex were received for postmortem examination. The clinical history was described as symptoms of mucopurulent nasal discharge, high to low grade fever, erosive stomatitis, dyspnoea and profuse watery diarrhoea followed by mortality of 35 goat kids within a week. The pathoanatomical lesions and immunohistochemical demonstration of PPRV antigen in lungs, intestine, spleen and lymph nodes confirmed PPR disease in goats. Grossly, five brain specimens showed moderate to severe leptomeningeal congestion during necropsy. Microscopically, brain sections showed leptomeningitis and nonsuppurative encephalitis characterized by vascular congestion, haemorrhages in the parenchyma, perivascular cuffing with mild to moderate mononuclear cells (mainly lymphocytes and few macrophages), focal to diffuse microgliosis, neuronal degeneration, satellitosis and neuronophagia. Immunolabelling of viral antigen was observed in the cytoplasm of neurons and glial cells. The RT-PCR amplification of N gene fragment also confirmed the presence of PPRV in the brain. The strong immunoreactivity of Caspase-3, Caspase-8 and comparatively lower expression of caspase-9 along with the absence of any reactivity for Apaf-1 antigen in the brain sections indicated the role of caspase dependent extrinsic pathway in inducing neuropathological changes. The presence of apoptotic neurons in the brain by TUNEL assay further confirmed the apoptosis and strong immunoreactivity of iNOS in neurons which suggested the generation of oxidative stress, that might have induced the apoptosis. The overall findings confirm the neurovirulence potential of PPR virus, via the extrinsic pathway of apoptosis, in natural cases of PPR disease in goat kids.


Asunto(s)
Caspasas/metabolismo , Enfermedades de las Cabras/enzimología , Peste de los Pequeños Rumiantes/enzimología , Animales , Apoptosis , Encéfalo/enzimología , Encéfalo/patología , Encéfalo/virología , Caspasas/genética , Femenino , Enfermedades de las Cabras/patología , Enfermedades de las Cabras/fisiopatología , Enfermedades de las Cabras/virología , Cabras , Pulmón/enzimología , Pulmón/patología , Pulmón/virología , Masculino , Neuropatología , Peste de los Pequeños Rumiantes/patología , Peste de los Pequeños Rumiantes/fisiopatología , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes/fisiología , Bazo/enzimología , Bazo/patología , Bazo/virología
11.
J Ethnopharmacol ; 248: 112279, 2020 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-31600562

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Virus de la Enfermedad de Newcastle/efectos de los fármacos , Virus de la Peste de los Pequeños Rumiantes/efectos de los fármacos , Extractos Vegetales/farmacología , Polyalthia , Animales , Chlorocebus aethiops , Virus de la Enfermedad de Newcastle/fisiología , Virus de la Peste de los Pequeños Rumiantes/fisiología , Hojas de la Planta , Células Vero , Internalización del Virus/efectos de los fármacos
12.
Viruses ; 11(12)2019 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-31817946

RESUMEN

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.


Asunto(s)
Camelus/virología , Especificidad del Huésped , Interacciones Huésped-Patógeno , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes/fisiología , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Biomarcadores , Biopsia , Bovinos , Femenino , Pruebas Hematológicas , Inmunohistoquímica , Masculino , Peste de los Pequeños Rumiantes/sangre , Peste de los Pequeños Rumiantes/patología
13.
Virus Res ; 274: 197774, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31606355

RESUMEN

Peste des petits ruminants (PPR) is an acute transboundary infectious viral disease affecting domestic and wild small ruminants' species besides camels reared in Africa, Asia and the Middle East. The virus is a serious paramount challenge to the sustainable agriculture advancement in the developing world. The disease outbreak was also detected for the first time in the European Union namely in Bulgaria at 2018. Therefore, the disease has lately been aimed for eradication with the purpose of worldwide clearance by 2030. Radically, the vaccines needed for effectively accomplishing this aim are presently convenient; however, the availableness of innovative modern vaccines to fulfill the desideratum for Differentiating between Infected and Vaccinated Animals (DIVA) may mitigate time spent and financial disbursement of serological monitoring and surveillance in the advanced levels for any disease obliteration campaign. We here highlight what is at the present time well-known about the virus and the different available diagnostic tools. Further, we interject on current updates and insights on several novel vaccines and on the possible current and prospective strategies to be applied for disease control.


Asunto(s)
Erradicación de la Enfermedad , Peste de los Pequeños Rumiantes/diagnóstico , Peste de los Pequeños Rumiantes/prevención & control , Virus de la Peste de los Pequeños Rumiantes , Animales , Técnicas de Laboratorio Clínico/veterinaria , Genoma Viral , Peste de los Pequeños Rumiantes/epidemiología , Virus de la Peste de los Pequeños Rumiantes/clasificación , Virus de la Peste de los Pequeños Rumiantes/inmunología , Virus de la Peste de los Pequeños Rumiantes/aislamiento & purificación , Virus de la Peste de los Pequeños Rumiantes/fisiología , Vacunación/veterinaria , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/clasificación , Vacunas Virales/administración & dosificación , Vacunas Virales/clasificación
14.
Viruses ; 11(8)2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31398809

RESUMEN

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.


Asunto(s)
Interacciones Huésped-Patógeno , Peste de los Pequeños Rumiantes/metabolismo , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes/fisiología , Receptores de Superficie Celular/metabolismo , Receptores Virales/metabolismo , Animales , Genoma Viral , Genómica/métodos , Especificidad del Huésped , Interacciones Huésped-Patógeno/inmunología , Inmunidad , Peste de los Pequeños Rumiantes/inmunología , Unión Proteica , Receptores de Superficie Celular/química , Receptores Virales/química , Transducción de Señal , Relación Estructura-Actividad , Proteínas Virales/química , Proteínas Virales/metabolismo
15.
Infect Genet Evol ; 75: 103981, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31369863

RESUMEN

Peste des petits ruminants virus (PPRV) is a morbillivirus which causes severe disease in ruminants. Since interferons (IFNs) serve as the important defense line against viral infection, we have investigated the roles of types I and III IFNs in PPRV infection in vitro. Upon PPRV infection, IFN-λ3 was strongly induced, while IFN-ß and IFN-λ2 were moderately induced at transcriptional level in human embryonic kidney 293 T (HEK293T) cells. Although the transcription of type I and III IFNs were triggered, the production of functional IFN products was not detected. Importantly, the replication of PPRV was strongly inhibited in HEK293T cells treated by the exogenous IFNs (IFN-α-2b, IFN-ß and IFN-λ3). Consistently, these IFNs significantly activate a panel of IFN-stimulated genes (ISGs). The inhibition of JAK-STAT pathway by JAK I inhibitor can abrogate the anti-PPRV activity of IFNs. Thus, our study shall contribute to better understanding of the complex PPRV-host interactions and provide rationale for therapeutic development of IFN-based treatment against PPRV infection.


Asunto(s)
Interferones/genética , Interferones/farmacología , Peste de los Pequeños Rumiantes/genética , Virus de la Peste de los Pequeños Rumiantes/fisiología , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Interacciones Microbiota-Huesped , Humanos , Quinasas Janus/genética , Peste de los Pequeños Rumiantes/tratamiento farmacológico , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes/efectos de los fármacos , Factores de Transcripción STAT/genética , Transducción de Señal/efectos de los fármacos , Replicación Viral/efectos de los fármacos
16.
Viruses ; 11(8)2019 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-31366072

RESUMEN

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.


Asunto(s)
Antígenos CD/genética , Fusión Celular , Glicoproteínas/genética , Interacciones Microbiota-Huesped/genética , Virus del Sarampión/genética , Virus de la Peste de los Pequeños Rumiantes/genética , Animales , Proteínas de la Cápside/genética , Línea Celular , Células Epiteliales/virología , Proteínas Ligadas a GPI/genética , Glicoproteínas/biosíntesis , Células HEK293 , Humanos , Virus del Sarampión/fisiología , Virus de la Peste de los Pequeños Rumiantes/fisiología , Ovinos , Proteínas Virales de Fusión/genética , Replicación Viral/genética
17.
J Virol ; 93(16)2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31167907

RESUMEN

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.


Asunto(s)
Interacciones Huésped-Patógeno , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/biosíntesis , Proteínas de la Nucleocápside/metabolismo , Peste de los Pequeños Rumiantes/metabolismo , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes/fisiología , Animales , Inmunomodulación , Interferón beta/genética , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Transporte de Proteínas , Activación Transcripcional
18.
Trop Anim Health Prod ; 51(7): 1807-1815, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31228086

RESUMEN

Mortality of domestic small ruminants caused by contagious caprine pleuropneumonia (CCPP) and Peste des petits ruminants (PPR) is frequently reported in Tanzania. A cross-sectional survey was conducted between June, 2016 and July, 2017 to identify risk factors for small ruminants exposure to Mycoplasma capricolum subsp. capripneumoniae (M. capripneumoniae), the causative agent of CCPP, and small ruminant morbillivirus (SRMV), the causative agent of PPR. Antibody detection was done using competitive enzyme-linked immunosorbent assays (cELISA); similarly, a semi-structured questionnaire was administered in flocks where serum samples were collected. Individual seropositivity for M. capripneumoniae was 6.5% (n = 676) and 4.2% (n = 285) in goats and sheep respectively, whereas SRMV was 28.6% in goats (n = 676) and 31.9% in sheep (n = 285). Multivariable analysis indicated that mixing of flocks was a risk factor for exposure to M. capripneumoniae (χ2 = 3.9, df = 1, p = 0.05) and SRMV (χ2 = 6.3, df = 1, p = 0.01) in goats. Age was a protective factor for SRMV seropositivity in both goats (χ2 = 7.4, df = 1, p = 0.006) and sheep (χ2 = 10.2, df = 1, p = 0.006). SRMV seropositivity in goats was also influenced by grazing in contact with wild animals (χ2 = 5.9, df = 1, p = 0.02) and taking animals to the animal markets (χ2 = 8.2, df = 1, p = 0.004). M. capripneumoniae and SRMV are influenced by several risk factors and their control needs concerted efforts between stakeholders, which may include community involvement in mandatory vaccination and animals' movement control.


Asunto(s)
Enfermedades de las Cabras/epidemiología , Mycoplasma capricolum/fisiología , Peste de los Pequeños Rumiantes/epidemiología , Virus de la Peste de los Pequeños Rumiantes/fisiología , Pleuroneumonía Contagiosa/epidemiología , Enfermedades de las Ovejas/epidemiología , Animales , Cabras , Factores de Riesgo , Ovinos , Tanzanía/epidemiología
19.
Virus Res ; 269: 197634, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31129173

RESUMEN

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 48 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.


Asunto(s)
Virus de la Peste de los Pequeños Rumiantes/fisiología , Proteínas Virales de Fusión/metabolismo , Ensamble de Virus , Replicación Viral , Animales , Transporte Biológico , Chlorocebus aethiops , Enfermedades de las Cabras/virología , Cabras/virología , Hemaglutininas Virales/metabolismo , Irak , Peste de los Pequeños Rumiantes/prevención & control , Virus de la Peste de los Pequeños Rumiantes/clasificación , Virus de la Peste de los Pequeños Rumiantes/inmunología , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Células Vero
20.
Res Vet Sci ; 124: 118-122, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30878633

RESUMEN

Peste des petits ruminants (PPR) is a major Transboundary animal disease (TADs) of sheep and goats in tropical regions caused by PPRV which can also infect cattle without any clinical signs but inducing seroconversion. However the epidemiological role of cattle in the maintenance and spread of the disease is not known. For the purposes of the present study, cattle were infected with a wild type candidate from each of the four lineages of PPRV and placed in separate boxes. Naive goats were then introduced into each specific box for the 30 days duration of the experiment. The results showed that no clinical signs of PPR were recorded in these infected cattle nor in the in-contact goats. The nasal and oral swabs remained negative. Serum from animals infected with three (3) of the wild type isolates of PPRV showed high percentage inhibition (PI % > 65%) in a cELISA. Only two animals out of three infected with the Nigeria 75/3 strain of lineage 2 (mild strain) had specific anti-PPR antibodies but with PI% values around the threshold of the test. Our findings suggest that cattle are dead-end hosts for PPRV and do not play an epidemiological role in the maintenance and spread of PPRV. In a PPR surveillance programme, cattle can serve as indicators of PPRV infection.


Asunto(s)
Enfermedades de los Bovinos/transmisión , Enfermedades de las Cabras/transmisión , Peste de los Pequeños Rumiantes/transmisión , Virus de la Peste de los Pequeños Rumiantes/fisiología , Animales , Bovinos , Enfermedades de los Bovinos/virología , Enfermedades de las Cabras/virología , Cabras , Peste de los Pequeños Rumiantes/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...