Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
PLoS Pathog ; 19(1): e1011136, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36716344

RESUMEN

African swine fever virus (ASFV) causes a devastating hemorrhagic disease with worldwide circulation and no widely available therapeutic prevention. The infectious particle has a multilayered architecture that is articulated upon an endoplasmic reticulum (ER)-derived inner envelope. This membrane acts as docking platform for the assembly of the outer icosahedral capsid and the underlying core shell, a bridging layer required for the formation of the central genome-containing nucleoid. While the details of outer capsid assembly are relatively well understood, those of core formation remain unclear. Here we report the functional characterization of pEP84R, a transmembrane polypeptide embedded in the inner envelope that surrounds the viral core. Using an ASFV recombinant inducibly expressing the EP84R gene, we show that absence of pEP84R results in the formation of non-infectious core-less icosahedral particles displaying a significant DNA-packaging defect. Concomitantly, aberrant core shell-like structures formed by co-assembly of viral polyproteins pp220 and pp62 are mistargeted to non-ER membranes, as also occurs when these are co-expressed in the absence of other viral proteins. Interestingly, co-expression of both polyproteins with pEP84R led to the formation of ER-targeted core shell-like assemblies and co-immunoprecipitation assays showed that pEP84R binds to the N-terminal region of pp220. Altogether, these results indicate that pEP84R plays a crucial role in core assembly by targeting the core shell polyproteins to the inner viral envelope, which enables subsequent genome packaging and nucleoid formation. These findings unveil a key regulatory mechanism for ASFV morphogenesis and identify a relevant novel target for the development of therapeutic tools against this re-emerging threat.


Asunto(s)
Virus de la Fiebre Porcina Africana , Fiebre Porcina Africana , Animales , Porcinos , Virus de la Fiebre Porcina Africana/genética , Virus de la Fiebre Porcina Africana/metabolismo , Ensamble de Virus , Proteínas Virales/genética , Proteínas Virales/metabolismo , Poliproteínas/metabolismo , Proteínas de la Membrana
2.
Proc Natl Acad Sci U S A ; 117(43): 26885-26894, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33046647

RESUMEN

Ectromelia virus (ECTV) causes mousepox, a surrogate mouse model for smallpox caused by variola virus in humans. Both orthopoxviruses encode tumor necrosis factor receptor (TNFR) homologs or viral TNFR (vTNFR). These homologs are termed cytokine response modifier (Crm) proteins, containing a TNF-binding domain and a chemokine-binding domain called smallpox virus-encoded chemokine receptor (SECRET) domain. ECTV encodes one vTNFR known as CrmD. Infection of ECTV-resistant C57BL/6 mice with a CrmD deletion mutant virus resulted in uniform mortality due to excessive TNF secretion and dysregulated inflammatory cytokine production. CrmD dampened pathology, leukocyte recruitment, and inflammatory cytokine production in lungs including TNF, IL-6, IL-10, and IFN-γ. Blockade of TNF, IL-6, or IL-10R function with monoclonal antibodies reduced lung pathology and provided 60 to 100% protection from otherwise lethal infection. IFN-γ caused lung pathology only when both the TNF-binding and SECRET domains were absent. Presence of the SECRET domain alone induced significantly higher levels of IL-1ß, IL-6, and IL-10, likely overcoming any protective effects that might have been afforded by anti-IFN-γ treatment. The use of TNF-deficient mice and those that express only membrane-associated but not secreted TNF revealed that CrmD is critically dependent on host TNF for its function. In vitro, recombinant Crm proteins from different orthopoxviruses bound to membrane-associated TNF and dampened inflammatory gene expression through reverse signaling. CrmD does not affect virus replication; however, it provides the host advantage by enabling survival. Host survival would facilitate virus spread, which would also provide an advantage to the virus.


Asunto(s)
Virus de la Ectromelia/fisiología , Interacciones Huésped-Patógeno , Receptores del Factor de Necrosis Tumoral/metabolismo , Infecciones del Sistema Respiratorio/virología , Proteínas Virales/metabolismo , Animales , Línea Celular , Chlorocebus aethiops , Femenino , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Infecciones del Sistema Respiratorio/patología , Carga Viral
3.
Cell Mol Life Sci ; 78(4): 1423-1444, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33084946

RESUMEN

Antiviral responses of interferons (IFNs) are crucial in the host immune response, playing a relevant role in controlling viralw infections. Three types of IFNs, type I (IFN-α, IFN-ß), II (IFN-γ) and III (IFN-λ), are classified according to their receptor usage, mode of induction, biological activity and amino acid sequence. Here, we provide a comprehensive review of type I IFN responses and different mechanisms that viruses employ to circumvent this response. In the first part, we will give an overview of the different induction and signaling cascades induced in the cell by IFN-I after virus encounter. Next, highlights of some of the mechanisms used by viruses to counteract the IFN induction will be described. And finally, we will address different mechanism used by viruses to interference with the IFN signaling cascade and the blockade of IFN induced antiviral activities.


Asunto(s)
Interacciones Huésped-Patógeno/genética , Inmunidad Innata/genética , Interferones/genética , Virosis/genética , Humanos , Evasión Inmune/genética , Interferón-alfa/genética , Interferón-alfa/inmunología , Interferones/clasificación , Interferones/inmunología , Transducción de Señal , Virosis/inmunología
4.
J Virol ; 92(23)2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30185597

RESUMEN

African swine fever virus (ASFV) is a large and complex DNA virus that causes a highly lethal swine disease for which there is no vaccine available. The ASFV particle, with an icosahedral multilayered structure, contains multiple polypeptides whose identity is largely unknown. Here, we analyzed by mass spectroscopy the protein composition of highly purified extracellular ASFV particles and performed immunoelectron microscopy to localize several of the detected proteins. The proteomic analysis identified 68 viral proteins, which account for 39% of the genome coding capacity. The ASFV proteome includes essentially all the previously described virion proteins and, interestingly, 44 newly identified virus-packaged polypeptides, half of which have an unknown function. A great proportion of the virion proteins are committed to the virus architecture, including two newly identified structural proteins, p5 and p8, which are derived from the core polyproteins pp220 and pp62, respectively. In addition, the virion contains a full complement of enzymes and factors involved in viral transcription, various enzymes implicated in DNA repair and protein modification, and some proteins concerned with virus entry and host defense evasion. Finally, 21 host proteins, many of them localized at the cell surface and related to the cortical actin cytoskeleton, were reproducibly detected in the ASFV particle. Immunoelectron microscopy strongly supports the suggestion that these host membrane-associated proteins are recruited during virus budding at actin-dependent membrane protrusions. Altogether, the results of this study provide a comprehensive model of the ASFV architecture that integrates both compositional and structural information.IMPORTANCE African swine fever virus causes a highly contagious and lethal disease of swine that currently affects many countries of sub-Saharan Africa, the Caucasus, the Russian Federation, and Eastern Europe and has very recently spread to China. Despite extensive research, effective vaccines or antiviral strategies are still lacking, and many basic questions on the molecular mechanisms underlying the infective cycle remain. One such gap regards the composition and structure of the infectious virus particle. In the study described in this report, we identified the set of viral and host proteins that compose the virion and determined or inferred the localization of many of them. This information significantly increases our understanding of the biological and structural features of an infectious African swine fever virus particle and will help direct future research efforts.


Asunto(s)
Virus de la Fiebre Porcina Africana/fisiología , Fiebre Porcina Africana/metabolismo , Poliproteínas/metabolismo , Proteoma/análisis , Proteínas Virales/metabolismo , Virión/metabolismo , Fiebre Porcina Africana/virología , Virus de la Fiebre Porcina Africana/ultraestructura , Secuencia de Aminoácidos , Animales , Chlorocebus aethiops , Microscopía Inmunoelectrónica , Porcinos , Células Vero , Virión/crecimiento & desarrollo
5.
Dis Aquat Organ ; 132(2): 151-156, 2019 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-30628581

RESUMEN

Lymphocystis disease, caused by the iridovirus lymphocystis disease virus (LCDV), is characterized by the appearance of tumour-like lesions on the skin of affected animals associated with several environmental factors and/or with stress due to the intensive culture conditions of fish farms. In a previous study, the genomes of a new LCDV species, LCDV-Sa, were detected, together with 2 previously unknown viruses, Sparus aurata papillomavirus 1 (SaPV1) and Sparus aurata polyomavirus 1 (SaPyV1). Gilthead seabream from 17 fish farms in Spain, Italy and Turkey were sampled between 2009 and 2015 to investigate the role of the newly described SaPV1 and SaPyV1 viruses in lymphocystis disease development. Our results show that in diseased fish, either or both of the new viruses are almost invariably detected together with LCDV (98%). In asymptomatic fish, these viruses were detected in a much lower percentage (28%) and mostly in concurrence with LCDV (24%). These data confirm the suspected association among the 3 different viruses during lymphocystis disease development in gilthead seabream and warrant future studies to establish their respective contributions.


Asunto(s)
Infecciones por Virus ADN , Enfermedades de los Peces , Poliomavirus , Dorada , Animales , Infecciones por Virus ADN/veterinaria , Italia , España , Turquía
6.
J Gen Virol ; 98(5): 1073-1079, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28555548

RESUMEN

Molluscum contagiosum virus (MCV) is the sole member of the Molluscipoxvirus genus and causes a highly prevalent human disease of the skin characterized by the formation of a variable number of lesions that can persist for prolonged periods of time. Two major genotypes, subtype 1 and subtype 2, are recognized, although currently only a single complete genomic sequence corresponding to MCV subtype 1 is available. Using next-generation sequencing techniques, we report the complete genomic sequence of four new MCV isolates, including the first one derived from a subtype 2. Comparisons suggest a relatively distant evolutionary split between both MCV subtypes. Further, our data illustrate concurrent circulation of distinct viruses within a population and reveal the existence of recombination events among them. These results help identify a set of MCV genes with potentially relevant roles in molluscum contagiosum epidemiology and pathogenesis.


Asunto(s)
Genoma Viral , Virus del Molusco Contagioso/clasificación , Virus del Molusco Contagioso/genética , Recombinación Genética , Niño , Análisis por Conglomerados , Voluntarios Sanos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Virus del Molusco Contagioso/crecimiento & desarrollo , Virus del Molusco Contagioso/aislamiento & purificación , Filogenia , Análisis de Secuencia de ADN , Homología de Secuencia
7.
J Virol ; 90(19): 8768-79, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27440877

RESUMEN

UNLABELLED: Lymphocystis disease is a geographically widespread disease affecting more than 150 different species of marine and freshwater fish. The disease, provoked by the iridovirus lymphocystis disease virus (LCDV), is characterized by the appearance of papillomalike lesions on the skin of affected animals that usually self-resolve over time. Development of the disease is usually associated with several environmental factors and, more frequently, with stress conditions provoked by the intensive culture conditions present in fish farms. In gilthead sea bream (Sparus aurata), an economically important cultured fish species in the Mediterranean area, a distinct LCDV has been identified but not yet completely characterized. We have used direct sequencing of the virome of lymphocystis lesions from affected S. aurata fish to obtain the complete genome of a new LCDV-Sa species that is the largest vertebrate iridovirus sequenced to date. Importantly, this approach allowed us to assemble the full-length circular genome sequence of two previously unknown viruses belonging to the papillomaviruses and polyomaviruses, termed Sparus aurata papillomavirus 1 (SaPV1) and Sparus aurata polyomavirus 1 (SaPyV1), respectively. Epidemiological surveys showed that lymphocystis disease was frequently associated with the concurrent appearance of one or both of the new viruses. SaPV1 has unique characteristics, such as an intron within the L1 gene, and as the first member of the Papillomaviridae family described in fish, provides evidence for a more ancient origin of this family than previously thought. IMPORTANCE: Lymphocystis disease affects marine and freshwater fish species worldwide. It is characterized by the appearance of papillomalike lesions on the skin that contain heavily enlarged cells (lymphocysts). The causative agent is the lymphocystis disease virus (LCDV), a large icosahedral virus of the family Iridoviridae In the Mediterranean area, the gilthead sea bream (Sparus aurata), an important farmed fish, is frequently affected. Using next-generation sequencing, we have identified within S. aurata lymphocystis lesions the concurrent presence of an additional LCDV species (LCDV-Sa) as well as two novel viruses. These are members of polyomavirus and papillomavirus families, and here we report them to be frequently associated with the presence of lymphocysts in affected fish. Because papillomaviruses have not been described in fish before, these findings support a more ancient origin of this virus family than previously thought and evolutionary implications are discussed.


Asunto(s)
Coinfección/veterinaria , Infecciones por Virus ADN/veterinaria , Enfermedades de los Peces/virología , Iridoviridae/aislamiento & purificación , Papillomaviridae/aislamiento & purificación , Poliomavirus/aislamiento & purificación , Dorada , Animales , Coinfección/patología , Coinfección/virología , Infecciones por Virus ADN/patología , Infecciones por Virus ADN/virología , ADN Viral/química , ADN Viral/genética , Enfermedades de los Peces/patología , Iridoviridae/clasificación , Iridoviridae/genética , Papillomaviridae/clasificación , Papillomaviridae/genética , Poliomavirus/clasificación , Poliomavirus/genética , Análisis de Secuencia de ADN
8.
J Biol Chem ; 290(26): 15973-84, 2015 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-25940088

RESUMEN

The blockade of tumor necrosis factor (TNF) by etanercept, a soluble version of the human TNF receptor 2 (hTNFR2), is a well established strategy to inhibit adverse TNF-mediated inflammatory responses in the clinic. A similar strategy is employed by poxviruses, encoding four viral TNF decoy receptor homologues (vTNFRs) named cytokine response modifier B (CrmB), CrmC, CrmD, and CrmE. These vTNFRs are differentially expressed by poxviral species, suggesting distinct immunomodulatory properties. Whereas the human variola virus and mouse ectromelia virus encode one vTNFR, the broad host range cowpox virus encodes all vTNFRs. We report the first comprehensive study of the functional and binding properties of these four vTNFRs, providing an explanation for their expression profile among different poxviruses. In addition, the vTNFRs activities were compared with the hTNFR2 used in the clinic. Interestingly, CrmB from variola virus, the causative agent of smallpox, is the most potent TNFR of those tested here including hTNFR2. Furthermore, we demonstrate a new immunomodulatory activity of vTNFRs, showing that CrmB and CrmD also inhibit the activity of lymphotoxin ß. Similarly, we report for the first time that the hTNFR2 blocks the biological activity of lymphotoxin ß. The characterization of vTNFRs optimized during virus-host evolution to modulate the host immune response provides relevant information about their potential role in pathogenesis and may be used to improve anti-inflammatory therapies based on soluble decoy TNFRs.


Asunto(s)
Virus de la Viruela Vacuna/metabolismo , Poxviridae/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/química , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Receptores Señuelo del Factor de Necrosis Tumoral/química , Receptores Señuelo del Factor de Necrosis Tumoral/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Virus de la Viruela Vacuna/química , Virus de la Viruela Vacuna/genética , Humanos , Linfotoxina beta/metabolismo , Ratones , Datos de Secuencia Molecular , Poxviridae/química , Poxviridae/genética , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Alineación de Secuencia , Receptores Señuelo del Factor de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/metabolismo , Proteínas Virales/genética
9.
J Virol ; 89(20): 10702-6, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26246565

RESUMEN

Amphibian-like ranaviruses include pathogens of fish, amphibians, and reptiles that have recently evolved from a fish-infecting ancestor. The molecular determinants of host range and virulence in this group are largely unknown, and currently fish infection models are lacking. We show that European sheatfish virus (ESV) can productively infect zebrafish, causing a lethal pathology, and describe a method for the generation of recombinant ESV, establishing a useful model for the study of fish ranavirus infections.


Asunto(s)
Infecciones por Virus ADN/veterinaria , Modelos Animales de Enfermedad , Enfermedades de los Peces/virología , Ranavirus/genética , Pez Cebra/virología , Animales , Secuencia de Bases , Infecciones por Virus ADN/patología , Infecciones por Virus ADN/virología , Enfermedades de los Peces/patología , Ingeniería Genética , Genotipo , Larva/virología , Datos de Secuencia Molecular , Filogenia , Ranavirus/clasificación , Ranavirus/patogenicidad , Virulencia
10.
J Gen Virol ; 96(10): 3118-3123, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26242179

RESUMEN

Poxviruses encode up to four different soluble TNF receptors, named cytokine response modifier B (CrmB), CrmC, CrmD and CrmE. These proteins mimic the extracellular domain of the cellular TNF receptors to bind and inhibit the activity of TNF and, in some cases, other TNF superfamily ligands. Most of these ligands are released after the enzymic cleavage of a membrane precursor. However, transmembrane TNF (tmTNF) is not only a precursor of soluble TNF but also exerts specific pro-inflammatory and immunological activities. Here, we report that viral TNF receptors bound and inhibited tmTNF and describe some interesting differences in their activity against the soluble cytokine. Thus, CrmE, which does not inhibit mouse soluble TNF, could block murine tmTNF-induced cytotoxicity. We propose that this anti-tmTNF effect should be taken into consideration when assessing the role of viral TNF decoy receptors in the pathogenesis of poxvirus.


Asunto(s)
Evasión Inmune , Poxviridae/inmunología , Poxviridae/fisiología , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Proteínas Virales/metabolismo , Animales , Línea Celular , Fibroblastos/inmunología , Fibroblastos/virología , Macrófagos/inmunología , Macrófagos/virología , Ratones
11.
Vet Res ; 46: 106, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26403184

RESUMEN

In 2010 a new Lagovirus related to rabbit haemorrhagic disease virus (RHDV) emerged in France and has since rapidly spread throughout domestic and wild rabbit populations of several European countries. The new virus, termed RHDV2, exhibits distinctive genetic, antigenic and pathogenic features. Notably, RHDV2 kills rabbits previously vaccinated with RHDV vaccines. Here we report for the first time the generation and characterization of RHDV2-specific virus-like particles (VLPs). Our results further confirmed the differential antigenic properties exhibited by RHDV and RHDV2, highlighting the need of using RHDV2-specific diagnostic assays to monitor the spread of this new virus.


Asunto(s)
Antígenos Virales/inmunología , Infecciones por Caliciviridae/veterinaria , Lagovirus/inmunología , Conejos , Animales , Infecciones por Caliciviridae/inmunología , Infecciones por Caliciviridae/virología , Virus de la Enfermedad Hemorrágica del Conejo/genética , Virus de la Enfermedad Hemorrágica del Conejo/inmunología , Lagovirus/genética , Filogenia , Análisis de Secuencia de ADN/veterinaria
12.
Antiviral Res ; : 105973, 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39168188

RESUMEN

African swine fever virus (ASFV) infection causes a frequently fatal disease in domestic swine that has affected more than 50 countries worldwide since 2021, with a major impact on animal welfare and economy. The development of effective vaccines or antivirals against this disease are urgently required for its effective control. Live detection of viral replication has been used as a tool for the screening and characterization of antiviral compounds in other dsDNA genome containing viruses. Here, we have adapted the ANCHOR fluorescent DNA labelling system to ASFV by constructing and characterizing a novel recombinant virus. We show that this virus is viable and effectively tags viral DNA replication sites, which can be detected and quantified in real time. Further, we have used high content cell microscopy to test the antiviral activity of bisbenzimide compounds and show that Hoechst 33342 has specific anti-ASFV activity. We expect this novel tool to be useful both in the further study of ASFV replication as in the screening of new specific antiviral compounds.

13.
J Virol ; 86(11): 6365-6, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22570241

RESUMEN

Viral diseases are an increasing threat to the thriving aquaculture industry worldwide. An emerging group of fish pathogens is formed by several ranaviruses, which have been isolated at different locations from freshwater and seawater fish species since 1985. We report the complete genome sequence of European sheatfish ranavirus (ESV), the first ranavirus isolated in Europe, which causes high mortality rates in infected sheatfish (Silurus glanis) and in other species. Analysis of the genome sequence shows that ESV belongs to the amphibian-like ranaviruses and is closely related to the epizootic hematopoietic necrosis virus (EHNV), a disease agent geographically confined to the Australian continent and notifiable to the World Organization for Animal Health.


Asunto(s)
Bagres/virología , ADN Viral/genética , Enfermedades de los Peces/virología , Genoma Viral , Ranavirus/genética , Análisis de Secuencia de ADN , Animales , ADN Viral/química , Europa (Continente) , Datos de Secuencia Molecular , Filogenia , Ranavirus/aislamiento & purificación , Homología de Secuencia
14.
J Virol ; 86(7): 3617-25, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22301140

RESUMEN

Worldwide amphibian population declines have been ascribed to global warming, increasing pollution levels, and other factors directly related to human activities. These factors may additionally be favoring the emergence of novel pathogens. In this report, we have determined the complete genome sequence of the emerging common midwife toad ranavirus (CMTV), which has caused fatal disease in several amphibian species across Europe. Phylogenetic and gene content analyses of the first complete genomic sequence from a ranavirus isolated in Europe show that CMTV is an amphibian-like ranavirus (ALRV). However, the CMTV genome structure is novel and represents an intermediate evolutionary stage between the two previously described ALRV groups. We find that CMTV clusters with several other ranaviruses isolated from different hosts and locations which might also be included in this novel ranavirus group. This work sheds light on the phylogenetic relationships within this complex group of emerging, disease-causing viruses.


Asunto(s)
Infecciones por Virus ADN/veterinaria , Evolución Molecular , Genoma Viral , Ranavirus/genética , Ranavirus/aislamiento & purificación , Salamandridae/virología , Animales , Secuencia de Bases , Infecciones por Virus ADN/virología , Europa (Continente) , Datos de Secuencia Molecular , Filogenia , Ranavirus/clasificación
15.
Virol J ; 10: 188, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23758704

RESUMEN

BACKGROUND: Lymphocystis disease virus (LCDV) is a large icosahedral dsDNA-containing virus of the Lymphocystivirus genus within the Iridoviridae family that can cause disease in more than 140 marine and freshwater fish species. While several isolates have been charcaterized and classified into distinct genotypes the complete genomic sequence is currently only available from two species, the LCDV-1, isolated from flounder (Platichtys flesus) in Europe and the LCDV-C, isolated from Japanese cultured flounder (Paralichthys olivaceus) in China. Analysis of the genome of LCDV-C showed it to encode a protein named LDVICp016 with similarities to the Tumour necrosis factor receptor (TNFR) superfamily with immunomodulatory potential. FINDINGS: We have expressed and purified the recombinant protein LDVICp016 and screened for potential interaction partners using surface plasmon resonance. Commercially available human and mouse members of the TNF superfamily (TNFSF), along with a representative set of fish-derived TNFSF were tested.We have found the LDVICp016 protein to be secreted and we have identified a second viral TNFR encoded by ORF 095 of the same virus. None of the 42 tested proteins were found to interact with LDVICp016. CONCLUSIONS: We show that LDVICp016 is a secreted protein belonging to the TNF receptor family that may be part of a larger gene family in Lymphocystiviruses. While the ligand of this protein remains unknown, possibly due to the species specific nature of this interaction, further investigations into the potential role of this protein in the blockade of immune responses in its fish host are required.


Asunto(s)
Iridoviridae/genética , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Animales , Peces , Humanos , Ratones , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
16.
FASEB J ; 25(6): 1960-71, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21372110

RESUMEN

Eradication of smallpox was accomplished 30 yr ago, but poxviral infections still represent a public health concern due to the potential release of variola virus or the emergence of zoonotic poxviruses, such as monkeypox virus. A critical determinant of poxvirus virulence is the inhibition of interferons (IFNs) by the virus-encoded type I IFN-binding protein (IFNα/ßBP). This immunomodulatory protein is secreted and has the unique property of interacting with the cell surface in order to prevent IFN-mediated antiviral responses. However, the mechanism of its attachment to the cell surface remains unknown. Using surface plasmon resonance and cell-binding assays, we report that the IFNα/ßBP from vaccinia virus, the smallpox vaccine, interacts with cell surface glycosaminoglycans (GAGs). Analysis of the contribution of different regions of the protein to cell surface binding demonstrated that clusters of basic residues in the first immunoglobulin domain mediate GAG interactions. Furthermore, mutation of the GAG-interaction motifs does not affect its IFN-binding and -blocking capacity. Functional conservation of GAG-binding sites is demonstrated for the IFNα/ßBP from variola and monkeypox viruses, extending our understanding of immune modulation by the most virulent human poxviruses. These results are relevant for the design of improved vaccines and intervention strategies.


Asunto(s)
Antivirales/farmacología , Regulación Viral de la Expresión Génica/fisiología , Glicosaminoglicanos/metabolismo , Interferones/metabolismo , Virus Vaccinia/metabolismo , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Células CHO , Clonación Molecular , Cricetinae , Cricetulus , Glicosaminoglicanos/genética , Células HeLa , Humanos , Interferones/antagonistas & inhibidores , Anotación de Secuencia Molecular , Mutación , Estructura Terciaria de Proteína , Virus Vaccinia/genética , Virus Vaccinia/inmunología , Proteínas Virales/genética
17.
FASEB J ; 24(5): 1479-88, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20019241

RESUMEN

Variola virus (VARV) caused smallpox, one of the most devastating human diseases and the first to be eradicated, but its deliberate release represents a dangerous threat. Virulent orthopoxviruses infecting humans, such as monkeypox virus (MPXV), could fill the niche left by smallpox eradication and the cessation of vaccination. However, immunomodulatory activities and virulence determinants of VARV and MPXV remain largely unexplored. We report the molecular characterization of the VARV- and MPXV-secreted type I interferon-binding proteins, which interact with the cell surface after secretion and prevent type I interferon responses. The proteins expressed in the baculovirus system have been purified, and their interferon-binding properties characterized by surface plasmon resonance. The ability of these proteins to inhibit a broad range of interferons was investigated to identify potential adaptation to the human immune system. Furthermore, we demonstrate by Western blot and activity assays the expression of the type I interferon inhibitor during VARV and MPXV infections. These findings are relevant for the design of new vaccines and therapeutics to smallpox and emergent virulent orthopoxviruses because the type I interferon-binding protein is a major virulence factor in animal models, vaccination with this protein induces protective immunity, and its neutralization prevents disease progression.


Asunto(s)
Interferón Tipo I/antagonistas & inhibidores , Monkeypox virus/inmunología , Mpox/inmunología , Viruela/inmunología , Virus de la Viruela/inmunología , Proteínas Virales/metabolismo , Factores de Virulencia/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Inmunomodulación , Monkeypox virus/metabolismo , Monkeypox virus/patogenicidad , Virus de la Viruela/metabolismo , Virus de la Viruela/patogenicidad , Vacunas Virales/inmunología
18.
Pharmaceuticals (Basel) ; 14(7)2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-34358101

RESUMEN

Foot and mouth disease is a highly contagious disease affecting cattle, sheep, and swine among other cloven-hoofed animals that imposes serious economic burden by its direct effects on farm productivity as well as on commerce of farmed produce. Vaccination using inactivated viral strains of the different serotypes is an effective protective measure, but has several drawbacks including a lack of cross protection and the perils associated with the large-scale growth of infectious virus. We have previously developed chimeric virus-like particles (VLPs) bearing an FMDV epitope which induced strong specific humoral responses in vaccinated pigs but conferred only partial protection against homologous challenge. While this and other FMD vaccines under development mostly rely on the induction of neutralizing responses, it is thought that induction of specific T-cell responses might improve both cross protective efficacy as well as duration of immunity. Therefore, we here describe the development of a recombinant adenovirus expressing the highly conserved nonstructural FMDV 3D protein as well as its capacity to induce specific T-cell responses in a murine model. We further describe the generation of an FMDV serotype C-specific chimeric VLP and analyze the immunogenicity of two different prime-boost strategies combining both elements in mice. This combination can effectively induce both humoral and cellular FMDV-specific responses eliciting high titers of ELISA and neutralizing antibodies anti-FMDV as well as a high frequency of IFNγ-secreting cells. These results provide the basis for further testing of this anti FMD vaccination strategy in cattle or pig, two of the most relevant natural host of this pathogen.

19.
Vaccines (Basel) ; 9(5)2021 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-34066934

RESUMEN

Currently there is a clear trend towards the establishment of virus-like particles (VLPs) as a powerful tool for vaccine development. VLPs are tunable nanoparticles that can be engineered to be used as platforms for multimeric display of foreign antigens. We have previously reported that VLPs derived from rabbit hemorrhagic disease virus (RHDV) constitute an excellent vaccine vector, capable of inducing specific protective immune responses against inserted heterologous T-cytotoxic and B-cell epitopes. Here, we evaluate the ability of chimeric RHDV VLPs to elicit immune response and protection against Foot-and-Mouth disease virus (FMDV), one of the most devastating livestock diseases. For this purpose, we generated a set of chimeric VLPs containing two FMDV-derived epitopes: a neutralizing B-cell epitope (VP1 (140-158)) and a T-cell epitope [3A (21-35)]. The epitopes were inserted joined or individually at two different locations within the RHDV capsid protein. The immunogenicity and protection potential of the chimeric VLPs were analyzed in the mouse and pig models. Herein we show that the RHDV engineered VLPs displaying FMDV-derived epitopes elicit a robust neutralizing immune response in mice and pigs, affording partial clinical protection against an FMDV challenge in pigs.

20.
Vaccines (Basel) ; 8(2)2020 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-32580486

RESUMEN

Members of the tumour necrosis factor (TNF) superfamily OX40L and CD70 and their receptors are costimulating signalling axes critical for adequate T cell activation in humans and mice but characterisation of these molecules in other species including ruminants is lacking. Here we cloned and expressed the predicted ovine orthologues of the receptors OX40 and CD27, as well as soluble recombinant forms of their potential ovine ligands, OaOX40L and OaCD70. Using biochemical and immunofluorescence analyses, we show that both signalling axes are functional in sheep. We show that oligomeric recombinant ligand constructs are able to induce signalling through their receptors on transfected cells. Recombinant defective human adenoviruses were constructed to express the soluble forms of OaOX40L and OaCD70. Both proteins were detected in the supernatant of adenovirus-infected cells and shown to activate NF-κB signalling pathway through their cognate receptor. These adenovirus-secreted OaOX40L and OaCD70 forms could also activate ovine T cell proliferation and enhance IFN-γ production in CD4+ and CD8+ T cells. Altogether, this study provides the first characterisation of the ovine costimulatory OX40L-OX40 and CD70-CD27 signalling axes, and indicates that their activation in vivo may be useful to enhance vaccination-induced immune responses in sheep and other ruminants.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA