Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
J Virol ; 98(4): e0014624, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38440983

RESUMEN

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.


Asunto(s)
Enfermedades de las Cabras , Cabras , Interacciones Huésped-Patógeno , Peste de los Pequeños Rumiantes , Virus de la Peste de los Pequeños Rumiantes , Activador de Plasminógeno de Tipo Uroquinasa , Proteínas Virales de Fusión , Animales , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína 58 DEAD Box/metabolismo , Enfermedades de las Cabras/inmunología , Enfermedades de las Cabras/metabolismo , Enfermedades de las Cabras/virología , Cabras/inmunología , Cabras/virología , Macrófagos Alveolares , Peste de los Pequeños Rumiantes/inmunología , Peste de los Pequeños Rumiantes/metabolismo , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes/crecimiento & desarrollo , Virus de la Peste de los Pequeños Rumiantes/inmunología , Virus de la Peste de los Pequeños Rumiantes/metabolismo , Unión Proteica , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Proteínas Virales de Fusión/metabolismo
2.
J Virol ; 96(18): e0124022, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-36094317

RESUMEN

Viruses have evolved numerous strategies to impair immunity so that they can replicate more efficiently. Among those, the immunosuppressive effects of morbillivirus infection can be particularly problematic, as they allow secondary infections to take hold in the host, worsening disease prognosis. In the present work, we hypothesized that the highly contagious morbillivirus peste des petits ruminants virus (PPRV) could target monocytes and dendritic cells (DC) to contribute to the immunosuppressive effects produced by the infection. Monocytes isolated from healthy sheep, a natural host of the disease, were able be infected by PPRV and this impaired the differentiation and phagocytic ability of immature monocyte-derived DC (MoDC). We also assessed PPRV capacity to infect differentiated MoDC. Ovine MoDC could be productively infected by PPRV, and this drastically reduced MoDC capacity to activate allogeneic T cell responses. Transcriptomic analysis of infected MoDC indicated that several tolerogenic DC signature genes were upregulated upon PPRV infection. Furthermore, PPRV-infected MoDC could impair the proliferative response of autologous CD4+ and CD8+ T cell to the mitogen concanavalin A (ConA), which indicated that DC targeting by the virus could promote immunosuppression. These results shed new light on the mechanisms employed by morbillivirus to suppress the host immune responses. IMPORTANCE Morbilliviruses pose a threat to global health given their high infectivity. The morbillivirus peste des petits ruminants virus (PPRV) severely affects small-ruminant-productivity and leads to important economic losses in communities that rely on these animals for subsistence. PPRV produces in the infected host a period of severe immunosuppression that opportunistic pathogens exploit, which worsens the course of the infection. The mechanisms of PPRV immunosuppression are not fully understood. In the present work, we demonstrate that PPRV can infect professional antigen-presenting cells called dendritic cells (DC) and disrupt their capacity to elicit an immune response. PPRV infection promoted a DC activation profile that favored the induction of tolerance instead of the activation of an antiviral immune response. These results shed new light on the mechanisms employed by morbilliviruses to suppress the immune responses.


Asunto(s)
Células Dendríticas , Activación de Linfocitos , Peste de los Pequeños Rumiantes , Virus de la Peste de los Pequeños Rumiantes , Animales , Antivirales , Diferenciación Celular , Concanavalina A/genética , Concanavalina A/inmunología , Células Dendríticas/citología , Células Dendríticas/virología , Cabras , Terapia de Inmunosupresión , Activación de Linfocitos/inmunología , Mitógenos/inmunología , Peste de los Pequeños Rumiantes/inmunología , Peste de los Pequeños Rumiantes/virología , Fenotipo , Ovinos , Linfocitos T/inmunología , Linfocitos T/virología
3.
Viruses ; 13(11)2021 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-34835126

RESUMEN

Across Africa, the Middle East, and Asia, peste des petits ruminants virus (PPRV) places a huge disease burden on agriculture, affecting, in particular, small ruminant production. The recent PPR outbreaks in Northern Africa, the European part of Turkey, and Bulgaria represent a significant threat to mainland Europe, as a source of disease. Although two safe and efficacious live attenuated vaccines (Sungri/96 and Nigeria/75/1) are available for the control of PPR, current serological tests do not enable the differentiation between naturally infected and vaccinated animals (DIVA). The vaccinated animals develop a full range of immune responses to viral proteins and, therefore, cannot be distinguished serologically from those that have recovered from a natural infection. This poses a serious problem for the post-vaccinal sero-surveillance during the ongoing PPR eradication program. Furthermore, during the latter stages of any eradication program, vaccination is only possible if the vaccine used is fully DIVA compliant. Using reverse genetics, we have developed two live attenuated PPR DIVA vaccines (Sungri/96 DIVA and Nigeria/75/1 DIVA), in which the C-terminal variable region of the PPRV N-protein has been replaced with dolphin morbillivirus (DMV). As a proof of principle, both the DIVA vaccines were evaluated in goats in pilot studies for safety and efficacy, and all the animals were clinically protected against the intranasal virulent virus challenge, similar to the parent vaccines. Furthermore, it is possible to differentiate between infected animals and vaccinated animals using two newly developed ELISAs. Therefore, these DIVA vaccines and associated tests can facilitate the sero-monitoring process and speed up the implementation of global PPR eradication through vaccination.


Asunto(s)
Enfermedades de los Animales , Peste de los Pequeños Rumiantes , Virus de la Peste de los Pequeños Rumiantes/inmunología , Rumiantes/virología , Vacunación/veterinaria , Vacunas Virales/inmunología , Enfermedades de los Animales/inmunología , Enfermedades de los Animales/prevención & control , Enfermedades de los Animales/virología , Animales , Peste de los Pequeños Rumiantes/inmunología , Peste de los Pequeños Rumiantes/prevención & control , Peste de los Pequeños Rumiantes/virología
4.
Front Immunol ; 12: 705539, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34594325

RESUMEN

The Morbillivirus peste des petits ruminants virus (PPRV) is the causal agent of a highly contagious disease that mostly affects sheep and goats and produces considerable losses in developing countries. Current PPRV control strategies rely on live-attenuated vaccines, which are not ideal, as they cannot differentiate infected from vaccinated animals (DIVA). Recombinant vector-based vaccines expressing viral subunits can provide an alternative to conventional vaccines, as they can be easily paired with DIVA diagnostic tools. In the present work, we used the bovine herpesvirus-4-based vector (BoHV-4-A) to deliver PPRV hemagglutinin H antigen (BoHV-4-A-PPRV-H-ΔTK). Vaccination with BoHV-4-A-PPRV-H-ΔTK protected sheep from virulent PPRV challenge and prevented virus shedding. Protection correlated with anti-PPRV IgGs, neutralizing antibodies and IFN-γ-producing cells induced by the vaccine. Detection of antibodies exclusively against H-PPRV in animal sera and not against other PPRV viral proteins such as F or N could serve as a DIVA diagnostic test when using BoHV-4-A-PPRV-H-ΔTK as vaccine. Our data indicate that BoHV-4-A-PPRV-H-ΔTK could be a promising new approach for PPRV eradication programs.


Asunto(s)
Vectores Genéticos , Herpesvirus Bovino 4 , Peste de los Pequeños Rumiantes/prevención & control , Virus de la Peste de los Pequeños Rumiantes , Enfermedades de las Ovejas/inmunología , Ovinos/inmunología , Proteínas Virales , Vacunas Virales , Animales , Chlorocebus aethiops , Perros , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Herpesvirus Bovino 4/genética , Herpesvirus Bovino 4/inmunología , Peste de los Pequeños Rumiantes/genética , Peste de los Pequeños Rumiantes/inmunología , Virus de la Peste de los Pequeños Rumiantes/genética , Virus de la Peste de los Pequeños Rumiantes/inmunología , Ovinos/virología , Enfermedades de las Ovejas/virología , Células Vero , Proteínas Virales/genética , Proteínas Virales/inmunología , Vacunas Virales/genética , Vacunas Virales/inmunología
5.
Front Immunol ; 12: 745315, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34671358

RESUMEN

Peste des petits ruminants (PPR) is an acute transboundary infectious viral disease of small ruminants, mainly sheep and goats. Host susceptibility varies considerably depending on the PPR virus (PPRV) strain, the host species and breed. The effect of strains with different levels of virulence on the modulation of the immune system has not been thoroughly compared in an experimental setting so far. In this study, we used a multi-omics approach to investigate the host cellular factors involved in different infection phenotypes. Peripheral blood mononuclear cells (PBMCs) from Saanen goats were activated with a T-cell mitogen and infected with PPRV strains of different virulence: Morocco 2008 (high virulence), Ivory Coast 1989 (low virulence) and Nigeria 75/1 (live attenuated vaccine strain). Our results showed that the highly virulent strain replicated better than the other two in PBMCs and rapidly induced cell death and a stronger inhibition of lymphocyte proliferation. However, all the strains affected lymphocyte proliferation and induced upregulation of key antiviral genes and proteins, meaning a classical antiviral response is orchestrated regardless of the virulence of the PPRV strain. On the other hand, the highly virulent strain induced stronger inflammatory responses and activated more genes related to lymphocyte migration and recruitment, and inflammatory processes. Both transcriptomic and proteomic approaches were successful in detecting viral and antiviral effectors under all conditions. The present work identified key immunological factors related to PPRV virulence in vitro.


Asunto(s)
Cabras/inmunología , Leucocitos Mononucleares/inmunología , Peste de los Pequeños Rumiantes/inmunología , Virus de la Peste de los Pequeños Rumiantes/patogenicidad , Virulencia/inmunología , Animales , Perfilación de la Expresión Génica , Cabras/virología , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes/inmunología , Proteómica
6.
Viruses ; 13(3)2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33807534

RESUMEN

The mitochondrial antiviral-signaling protein (MAVS, also known as VISA, IPS-1, or CARDIF) plays an essential role in the type I interferon (IFN) response and in retinoic acid-inducible gene I (RIG-I) mediated antiviral innate immunity in mammals. In this study, the caprine MAVS gene (caMAVS, 1566 bp) was identified and cloned. The caMAVS shares the highest amino acid similarity (98.1%) with the predicted sheep MAVS. Confocal microscopy analysis of partial deletion mutants of caMAVS revealed that the transmembrane and the so-called Non-Characterized domains are indispensable for intracellular localization to mitochondria. Overexpression of caMAVS in caprine endometrial epithelial cells up-regulated the mRNA levels of caprine interferon-stimulated genes. We concluded that caprine MAVS mediates the activation of the type I IFN pathway. We further demonstrated that both the CARD-like domain and the transmembrane domain of caMAVS were essential for the activation of the IFN-ß promotor. The interaction between caMAVS and caprine RIG-I and the vital role of the CARD and NC domain in this interaction was demonstrated by co-immunoprecipitation. Upon infection with the Peste des Petits Ruminants Virus (PPRV, genus Morbillivirus), the level of MAVS was greatly reduced. This reduction was prevented by the addition of the proteasome inhibitor MG132. Moreover, we found that viral protein V could interact and colocalize with MAVS. Together, we identified caMAVS as a RIG-I interactive protein involved in the activation of type I IFN pathways in caprine cells and as a target for PPRV immune evasion.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Inductores de Interferón/inmunología , Peste de los Pequeños Rumiantes/inmunología , Virus de la Peste de los Pequeños Rumiantes/inmunología , Animales , Chlorocebus aethiops , Células Epiteliales , Cabras , Células HEK293 , Humanos , Interferón Tipo I/inmunología , Células Vero
7.
Front Immunol ; 12: 630307, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33912160

RESUMEN

The study was aimed at developing an accessible laboratory animal model to elucidate protective and pathological roles of immune mediators during Peste des petits ruminants virus (PPRV) infection. It is because of the critical roles of type I IFNs in anti-viral defense, we assessed the susceptibility of IFN receptor knock out (IFNR KO) mice to PPRV infection. IFNR KO mice were exceedingly susceptible to the infection but WT animals efficiently controlled PPRV. Accordingly, the PPRV infected IFNR KO mice gradually reduced their body weights and succumbed to the infection within 10 days irrespective of the dose and route of infection. The lower infecting doses predominantly induced immunopathological lesions. The viral antigens as well as the replicating PPRV were abundantly present in most of the critical organs such as brain, lungs, heart and kidneys of IFNR KO mice infected with high dose of the virus. Neutrophils and macrophages transported the replicating virus to central nervous system (CNS) and contributed to pathology while the elevated NK and T cell responses directly correlated with the resolution of PPRV infection in WT animals. Using an array of fluorescently labeled H-2Kb tetramers, we discovered four immunogenic epitopes of PPRV. The PPRV-peptides interacted well with H-2Kb in acellular and cellular assay as well as expanded the virus-specific CD8+ T cells in immunized or infected mice. Adoptively transferred CD8+ T cells helped control PPRV in infected mice. Our study therefore established and employed a mouse model for investigating the pathogenesis of PPRV. The model could be useful for elucidating the contribution of immune cells in disease progression as well as to test anti-viral agents.


Asunto(s)
Peste de los Pequeños Rumiantes/inmunología , Animales , Encéfalo/virología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Antígenos H-2/inmunología , Inmunidad Innata , Inmunización , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Peste de los Pequeños Rumiantes/mortalidad , Peste de los Pequeños Rumiantes/patología , Virus de la Peste de los Pequeños Rumiantes/inmunología , Receptores de Interferón/fisiología , Vacunas Virales/inmunología
8.
J Immunoassay Immunochem ; 42(4): 424-443, 2021 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-33724901

RESUMEN

This study reports the influence of peste des petits ruminants (PPR) vaccination on the clinico-pathological outcomes of PPR in the face of an outbreak. Twenty-two West African dwarf goats procured for a different study started showing early signs of PPR during acclimatization. In response, PPR vaccine was administered either intranasally with phytogenic mucoadhesive gum (Group A; n = 6) or without gum (Group B; n = 6); subcutaneously (Group C; n = 6) or not vaccinated (Group D; n = 4) and studied for 21 days. The clinical scores, hematology, serology and pathology scores were evaluated. Clinical signs of PPR were present in all groups, presenting a percentage mortality of 33%; 33%; 64% and 100% for Groups A, B, C, and D, respectively. Polycythemia and mild leukopenia were observed in all groups, and all animals were seropositive by day 7 post-vaccination. The lung consolidation scores were low in Groups A and B, compared to Group C. Histopathological lesions consistent with PPR was observed in the lymphoid organs, gastrointestinal tract, and lungs with the presence of PPR antigen as detected by immunohistochemistry. The findings suggest that intranasal vaccination with or without mucoadhesive gum may influence the outcome of PPR infection more than the subcutaneous route in the face of an outbreak.


Asunto(s)
Sistemas de Liberación de Medicamentos , Peste de los Pequeños Rumiantes/inmunología , Vacunas Virales/inmunología , Administración Intranasal , Animales , Encía/inmunología , Cabras , Inyecciones Subcutáneas , Masculino , Virus de la Peste de los Pequeños Rumiantes/inmunología , Polímeros/administración & dosificación , Resultado del Tratamiento , Vacunas Virales/administración & dosificación
9.
J Immunoassay Immunochem ; 42(1): 82-94, 2021 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-32970525

RESUMEN

Peste des petits ruminants (PPR) in Africa continues to defy conventional vaccinational approaches aimed at its control. There is need for route modification and immunopotentiation of the current vaccination methods, using easily affordable materials. This study evaluates the immunomodulatory potential of Irvingia gabonensis (IG) seed gum extract for intranasal PPR vaccination in goats using attenuated Nigeria 75/1 PPR vaccine. Twenty West African dwarf goats were divided into four groups (n=5). Group 1 was vaccinated intranasally using IG gum as vehicle; Group 2 was vaccinated intranasally without the gum; Group 3 via subcutaneous injection while Group 4 was not vaccinated. Hematology and Serum IgG levels were assessed weekly for 28 days post vaccination (dpv). H-PPR bELISA detected antibodies against PPR by 7th dpv, peaking by 21st dpv with mean percentage inhibitions of 78.2%; 69.6%; 87.0% and 0% in Groups 1, 2, 3 and 4, respectively. Also, significantly lower neutrophil to lymphocyte ratio (P<0.05) were observed by 14th dpv to 28th dpv in the vaccinated groups. The findings of this study show that the use of I. gabonensis seed gum extract for mucoadhesive intranasal PPR vaccine delivery has an immunomodulatory effect on the systemic immune response following PPR intranasal vaccine administration.


Asunto(s)
Celulosa/química , Sistemas de Liberación de Medicamentos , Inmunidad Humoral/inmunología , Peste de los Pequeños Rumiantes/inmunología , Virus de la Peste de los Pequeños Rumiantes/inmunología , Vacunas Virales/inmunología , Administración Intranasal , Animales , Celulosa/inmunología , Cabras , Hematócrito , Hemoglobinas/análisis , Recuento de Linfocitos , Polímeros/química , Polímeros/aislamiento & purificación , Vacunación , Vacunas Virales/administración & dosificación
10.
Transbound Emerg Dis ; 68(6): 3320-3334, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33222411

RESUMEN

Peste des petits ruminants (PPR) is a severe disease of goats and sheep that is widespread in Africa, the Middle East and Asia. The disease is caused by peste des petits ruminants virus (PPRV); cell culture-attenuated strains of PPRV have been shown, both experimentally and by extensive use in the field, to be effective vaccines and are widely used. We have previously demonstrated that these vaccines elicit both serological (PPRV-specific antibody) and cell-based (PPRV-specific CD4+ and CD8+ T cells) immune responses. However, it is not known which of these responses are required for protection from PPRV, information that would be useful in the evaluation of new vaccines that are being developed to provide the capability to differentiate infected and vaccinated animals (DIVA capability). To begin to address this issue, we have used a complement-fixing monoclonal antibody recognizing caprine CD8 to deplete >99.9% of circulating CD8+ T cells from vaccinated goats. Animals were then infected with wild-type PPRV. Despite the absence of the CD8+ T-cell component of the vaccine-induced immune response, the vaccinated animals were almost fully protected, showing no pyrexia or viraemia, and almost no clinical signs. These data suggest that a virus-specific CD8+ T-cell response is not critical for protection against PPRV and that virus-specific antibody and/or CD4+ T cells are the main mediators of protection. We have also shown that the leucopenia caused by infection with wild-type PPRV affects all major classes of circulating leucocytes.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Enfermedades de las Cabras , Peste de los Pequeños Rumiantes , Vacunas Virales , Animales , Anticuerpos Antivirales , Enfermedades de las Cabras/inmunología , Enfermedades de las Cabras/prevención & control , Cabras , Peste de los Pequeños Rumiantes/inmunología , Peste de los Pequeños Rumiantes/prevención & control , Virus de la Peste de los Pequeños Rumiantes
11.
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
12.
Transbound Emerg Dis ; 67(6): 2881-2891, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32502324

RESUMEN

Peste des petits ruminants (PPR) is a fatal disease of small ruminants which has spread rapidly to previously PPR-free countries in recent decades, causing enormous economic losses in the affected regions. Here, two newly emerged PPR virus (PPRV) isolates from India and from the Middle East were tested in an animal trial to analyse their pathogenesis, and to evaluate serological and molecular detection methods. Animals infected with the two different PPRV isolates showed marked differences in clinical manifestation and scoring. The PPRV isolate from India was less virulent than the virus from the Middle East. Commercially available rapid detection methods for PPRV antigen (two Lateral Flow Devices (LFDs) and one antigen ELISA) were evaluated in comparison with a nucleic acid detection method. For this purpose, ocular and nasal swabs were used. Due to the easy non-invasive sampling, faecal samples were also analysed. For all rapid antigen detection methods, a high specificity of 100% was observed independent of the sample matrix and dilution buffers used. Both antigen ELISA and LFD tests showed highest sensitivities for nasal swabs. Here, the detection rate of the antigen ELISA, the LFD-PESTE-TEST and the LFD-ID Rapid-Test was 78%, 75% and 78%, respectively. Ocular swabs were less suitable for antigen detection of PPRV. These results reflect the increased viral load in nasal swabs of PPRV infected goats compared to ocular swabs. The faecal samples were the least suitable for antigen detection. In conclusion, nasal swab samples are the first choice for the antigen and genome detection of PPRV. Nevertheless, based on the excellent diagnostic specificity of the rapid tests, positive results generated with other sample matrices are solid. In contrast, negative test results can be caused on the reduced analytical sensitivity of the rapid antigen tests and must be treated with caution.


Asunto(s)
Antígenos Virales/inmunología , Enfermedades de las Cabras/diagnóstico , Peste de los Pequeños Rumiantes/diagnóstico , Virus de la Peste de los Pequeños Rumiantes/inmunología , Animales , Ensayo de Inmunoadsorción Enzimática/veterinaria , Heces/virología , Enfermedades de las Cabras/inmunología , Enfermedades de las Cabras/virología , Cabras , India , Masculino , Mucosa Nasal/virología , Peste de los Pequeños Rumiantes/inmunología , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes/aislamiento & purificación , Pruebas de Precipitina/veterinaria
13.
Gene ; 754: 144858, 2020 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-32531455

RESUMEN

Not only are autophagy-related (ATG) proteins the essential orchestrators of the autophagy machinery, but also they regulate many other cellular pathways. Here, we demonstrated that ATG13 exerted an obviously antiviral activity against the infection of peste des petits ruminants virus (PPRV) in cell culture model. We found that PPRV infection or the treatment with interferon (IFN) against PPRV infection significantly induced ATG13 expression. Mechanistically, ATG13 stimulated interferon expression and the subsequent activation of the JAK-STAT cascade. These activations triggered the transcription of interferon-stimulated genes (ISGs) to exert antiviral activity. Conversely, the loss of ATG13 significantly attenuated the potency of RIG-IN in activating IFN responses. In summary, we have demonstrated that basal ATG13 was involved in host antiviral activities against PPRV infection and the over-expression of ATG13 activated IFN production to inhibit PPRV replication in an unconventional fashion.


Asunto(s)
Antivirales/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Interferones/farmacología , Peste de los Pequeños Rumiantes/tratamiento farmacológico , Virus de la Peste de los Pequeños Rumiantes/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Autofagia , Proteínas Relacionadas con la Autofagia , Células HEK293 , Humanos , Peste de los Pequeños Rumiantes/inmunología , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes/inmunología , Transducción de Señal
14.
J Immunoassay Immunochem ; 41(3): 311-321, 2020 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-32119592

RESUMEN

A study was conducted to evaluate mucoadhesive property and immunomodulatory effect of phytogenic gums from Boswellia frereana, Boswellia carteri andCommiphora myrrha on intranasal Peste des petits ruminants (PPR) vaccination in goats and sheep in an ex-vivo and in-vivo situations. Plant gums were purified, dried and compressed into 500gm tablets. Modified shear stress measurement technique was used on freshly excised trachea and intestine tissues of goat to measure peak adhesion time. Forty eight animals (24 goats and 24 sheep) were divided into eight groups (of 3 goats and 3 sheep) and immunized intranasally with gum-vaccine combinations in two ratios (1:1, 1:2). Antibody against PPR virus was measured on day 14, 28, 42 and 56 post vaccination using H-based PPR bELISA. The peak adhesion time of the different gums was transient. PPR virus antibodies were detected in all immunized goats and sheep but not in unvaccinated control. The best percentage inhibition was recorded for Boswellia carteri-vaccine combination group at a ratio of 1:1. Administration of Boswellia carteri-PPR vaccine combination through intranasal or subcutaneous route, elicited similar antibody titre, implying that the intranasal route may be used as a non-invasive alternative delivery in PPR vaccination of small ruminants.


Asunto(s)
Anticuerpos Antivirales/inmunología , Boswellia/química , Boswellia/inmunología , Peste de los Pequeños Rumiantes/inmunología , Resinas de Plantas/administración & dosificación , Resinas de Plantas/farmacología , Vacunación , Vacunas Virales/inmunología , Adhesividad , Administración Intranasal , Animales , Anticuerpos Antivirales/administración & dosificación , Anticuerpos Antivirales/aislamiento & purificación , Mucosa Gástrica , Cabras , Peste de los Pequeños Rumiantes/terapia , Virus de la Peste de los Pequeños Rumiantes/inmunología , Resinas de Plantas/aislamiento & purificación , Ovinos , Vacunas Virales/administración & dosificación , Vacunas Virales/aislamiento & purificación
15.
Viruses ; 12(3)2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-32183412

RESUMEN

Peste des Petits Ruminants (PPR) is a highly contagious viral disease of both domestic (goats and sheep) and wild ruminants. Caused by a morbillivirus, that belongs to the family Paramyxoviridae. The disease is clinically and pathologically similar to rinderpest of cattle and human measles. PPR is one of the most economically devastating viral diseases of small ruminants. In April 2015, the Food and Agriculture Organization of the United Nations (FAO) and the World Organisation for Animal Health (OIE) launched the PPR Global Control and Eradication Strategy (PPR GCES) with the vision for global eradication by 2030. There is a strong and lasting international consensus to eradicate the disease in order to protect the livelihoods of the world's poorest populations. As with any disease, eradication is feasible when, policy, scientific and technical challenges are addressed. Ten majors challenges are described in this paper namely: understanding small ruminant production, facilitating research to support eradication, refining laboratory testing, improving epidemiological understanding of the virus, defining infection of wildlife and other species, optimizing vaccine delivery and novel vaccines, developing better control of animal movement, heightening serological monitoring, understanding socio-economic impact, and garnering funding and political will.


Asunto(s)
Peste de los Pequeños Rumiantes/prevención & control , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes , Animales , Bovinos , Erradicación de la Enfermedad , Cabras , Peste de los Pequeños Rumiantes/inmunología , Rumiantes/virología , Ovinos , Vacunas Virales/inmunología , Organización Mundial de la Salud
16.
J Virol ; 94(6)2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31896600

RESUMEN

Differentiating infected from vaccinated animals (DIVA) strategies have been central enabling techniques in several successful viral disease elimination programs. However, owing to their long and uncertain development process, no DIVA-compatible vaccines are available for many important diseases. We report herein a new DIVA strategy based on hybrid protein-peptide microarrays which can theoretically work with any vaccine. Leading from our findings from peste des petits ruminants (PPR) virus, we found 4 epitope-containing short peptides (ECSPs) which have distinct IgG serodynamics: anti-ECSP IgGs only exist for 10 to 60 days postvaccination (dpv), while anti-protein IgGs remained at high levels for >1,000 dpv. These data enabled the design of a DIVA diagnostic microarray containing 4 ECSPs and 3 proteins, which, unlike competitive enzyme-linked immunosorbent assay (cELISA) and virus neutralization tests (VNTs), enables ongoing monitoring of serological differences between vaccinated individuals and individuals exposed to the pathogen. For 25 goats after 60 dpv, 13 were detected with positive anti-ECSP IgGs, indicating recent infections in vaccinated goat herds. These DIVA diagnostic microarrays will almost certainly facilitate eradication programs for (re)emerging pathogens and zoonoses.IMPORTANCE Outbreaks of infectious diseases caused by viruses, such as pseudorabies (PR), foot-and-mouth disease (FMD), and PPR viruses, led to economic losses reaching billions of dollars. Both PR and FMD were eliminated in several countries via large-scale vaccination programs using DIVA-compatible vaccines, which lack the gE protein and nonstructural proteins, respectively. However, there are still extensive challenges facing the development and deployment of DIVA-compatible vaccines because they are time-consuming and full of uncertainty. Further, the negative marker strategy used for DIVA-compatible vaccines is no longer functional for live-attenuated vaccines. To avoid these disadvantageous scenarios, a new strategy is desired. Here, we made the exciting discovery that different IgG serodynamics can be monitored when using protein-based assays versus arrays comprising ECSPs. This DIVA microarray strategy should, in theory, work for any vaccine.


Asunto(s)
Anticuerpos Antivirales/inmunología , Epítopos/química , Inmunoglobulina G/inmunología , Péptidos/química , Peste de los Pequeños Rumiantes/inmunología , Virus de la Peste de los Pequeños Rumiantes/inmunología , Análisis por Matrices de Proteínas , Vacunación , Animales , Fiebre Aftosa/inmunología , Fiebre Aftosa/prevención & control , Cabras , Peste de los Pequeños Rumiantes/prevención & control , Seudorrabia/inmunología , Seudorrabia/prevención & control , Vacunas Virales/inmunología
17.
Autophagy ; 16(5): 842-861, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31318632

RESUMEN

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.


Asunto(s)
Autofagosomas/metabolismo , Autofagia/fisiología , Lisosomas/metabolismo , Peste de los Pequeños Rumiantes/metabolismo , Animales , Interferones/metabolismo , Peste de los Pequeños Rumiantes/inmunología , Virus de la Peste de los Pequeños Rumiantes/genética , Virus de la Peste de los Pequeños Rumiantes/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/fisiología , Replicación Viral/fisiología
18.
Prev Vet Med ; 174: 104850, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31794918

RESUMEN

Vaccination is the main tool for control of peste des petits ruminants (PPR) because of the availability of effective and safe vaccines that provide long lasting protection. However vaccination campaigns may not always provide sufficient herd immunity needed to prevent disease outbreaks because of logistic problems with vaccination such as inappropriate cold chain and vaccine delivery methods, and the rapid population turnover of small ruminants. This study was carried out to assess post-vaccination herd immunity against PPR and inter-vaccination population turnover in small ruminant flocks in Metema district, northwest Ethiopia where frequent PPR outbreaks occur despite regular vaccination. A total of 412 serum samples were collected from selected small ruminants in 72 flocks (average flock size of 33.4 and standard deviation of 30) above three months of age in three kebeles immediately before a vaccination program. One month after the vaccination using freeze dried live attenuated vaccine, 359 serum samples were collected from randomly selected small ruminants in the same flocks. The collected serum samples were analyzed to determine the seropositivity using a monoclonal antibody-based C-ELISA. The pre-vaccination seropositivity of 72.3% (95% CI: 67.8-76.4) increased to 93.9% (95% CI: 90.9-95.9) post-vaccination (P < 0.001). The observed seropositivity following vaccination was above the recommended herd immunity threshold (80%) required to reduce the transmission of infection in the population sufficient to eliminate virus. A survey of sampled flocks six months post-sampling indicated only 68% of animals were still present in these flocks. This population turnover reduces the herd immunity to about 64% which is below the required threshold for control. The high level of herd immunity achieved post-vaccination indicates good vaccine quality, cold chain maintenance and effective vaccine delivery in the district's vaccination campaigns. The decrease in herd immunity associated with population turnover and annual vaccination intervals represents a challenge to effective control and suggests changes to the timing or frequency of the vaccination is required.


Asunto(s)
Enfermedades de las Cabras/prevención & control , Inmunidad Colectiva , Peste de los Pequeños Rumiantes/prevención & control , Enfermedades de las Ovejas/prevención & control , Vacunación/veterinaria , Animales , Etiopía , Enfermedades de las Cabras/inmunología , Cabras , Peste de los Pequeños Rumiantes/inmunología , Dinámica Poblacional , Ovinos , Enfermedades de las Ovejas/inmunología
19.
Viruses ; 11(11)2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31694166

RESUMEN

Peste des petits ruminants (PPR) is one of the highly contagious transboundary viral diseases of small ruminants. Host microRNA (miRNA) expression patterns may change in response to virus infection, and it mainly works as a post-transcriptional moderator in gene expression and affects viral pathogenesis and replication. In this study, the change of miRNA expression profile in peripheral blood lymphocyte (PBMC) from sheep inoculated with PPR vaccine virus in vivo as well as primary sheep testicular (ST) cells inoculated with PPR vaccine virus in vitro were determined via deep sequencing technology. In PBMC cells, 373 and 115 differentially expressed miRNAs (DEmiRNAs) were identified 3 days and 5 days post inoculated (dpi), respectively. While, 575 DEmiRNAs were identified when comparing miRNA profiles on 5 dpi with 3 dpi. Some of the DEmiRNAs were found to change significantly via time-course during PPR vaccine virus inoculated. Similarly, in ST cells, 136 DEmiRNAs were identified at 3 dpi in comparison with mock-inoculation. A total of 12 DEmiRNAs were validated by real-time quantitative PCR (RT-qPCR). The oar-miR-150, oar-miR-370-3p and oar-miR-411b-3p were found common differentially expressed in both PPR vaccine virus-inoculated PBMC cells and ST cells. Targets prediction and functional analysis of the DEmiRNAs uncovered mainly gathering in antigen processing and presentation pathways, protein processing in endoplasmic reticulum pathways and cell adhesion molecules pathways. Our study supplies information about the DEmiRNAs in PPR vaccine virus-inoculated PBMC cells and ST cells, and provides clues for further understanding the function of miRNAs in PPR vaccine virus replication.


Asunto(s)
Leucocitos Mononucleares/metabolismo , MicroARNs/genética , Virus de la Peste de los Pequeños Rumiantes/inmunología , Vacunas Virales/inmunología , Animales , Células Cultivadas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Leucocitos Mononucleares/virología , MicroARNs/metabolismo , Peste de los Pequeños Rumiantes/genética , Peste de los Pequeños Rumiantes/inmunología , Virus de la Peste de los Pequeños Rumiantes/genética , ARN Viral/metabolismo , Ovinos , Vacunación/veterinaria , Vacunas Virales/administración & dosificación
20.
Viruses ; 11(10)2019 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-31590353

RESUMEN

Peste des petits ruminants is a highly contagious acute or subacute disease of small ruminants caused by the peste des petits ruminants virus (PPRV), and it is responsible for significant economic losses in animal husbandry. Vaccination represents the most effective means of controlling this disease, with virus-like particle (VLP) vaccines offering promising vaccine candidates. In this study, a PPRV VLP-based vaccine was developed using a baculovirus expression system, allowing for the simultaneous expression of the PPRV matrix (M), hemagglutinin (H), fusion (F) and nucleocapsid (N) proteins in insect cells. Immunization of mice and goats with PPRV VLPs elicited a robust neutralization response and a potent cellular immune response. Mouse studies demonstrated that VLPs induced a more robust IFN-γ response in CD4+ and CD8+ T cells than PPRV Nigeria 75/1 and recruited and/or activated more B cells and dendritic cells in inguinal lymph nodes. In addition, PPRV VLPs induced a strong Th1 class response in mice, as indicated by a high IgG2a to IgG1 ratio. Goat studies demonstrated that PPRV VLPs can induce the production of antibodies specific for F and H proteins and can also stimulate the production of virus neutralizing antibodies to the same magnitude as the PPRV Nigeria 75/1 vaccine. Higher amounts of IFN-γ in VLP-immunized animal serum suggested that VLPs also elicited a cellular immune response in goats. These results demonstrated that VLPs elicit a potent immune response against PPRV infection in small ruminants, making PPRV VLPs a potential candidate for PPRV vaccine development.


Asunto(s)
Cabras/inmunología , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Peste de los Pequeños Rumiantes/inmunología , Virus de la Peste de los Pequeños Rumiantes/inmunología , Animales , Anticuerpos Antivirales , Baculoviridae/genética , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos/inmunología , Citocinas/sangre , Modelos Animales de Enfermedad , Femenino , Enfermedades de las Cabras/virología , Inmunización , Masculino , Ratones , Ratones Endogámicos BALB C , Peste de los Pequeños Rumiantes/prevención & control , Virus de la Peste de los Pequeños Rumiantes/genética , Células Sf9 , Vacunas Virales/inmunología , Virión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...