85 results on '"Noemí Sevilla"'
Search Results
2. Comprehensive immune profiling reveals that Orbivirus infection activates immune checkpoints during acute T cell immunosuppression
- Author
-
Andrés Louloudes-Lázaro, José M. Rojas, Isabel García-García, Daniel Rodríguez-Martín, Esther Morel, Verónica Martín, and Noemí Sevilla
- Subjects
Orbivirus ,T cells ,PD-1/PD-L1 checkpoint ,transcriptomic ,monocytes ,B cells ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Bluetongue virus (BTV) is an arbovirus transmitted by the bite of infected Culicoides midges that affects domestic and wild ruminants producing great economic losses. The infection induces an IFN response, followed by an adaptive immune response that is essential in disease clearance. BTV can nonetheless impair IFN and humoral responses. The main goal of this study was to gain a more detailed understanding of BTV pathogenesis and its effects on immune cell populations. To this end, we combined flow cytometry and transcriptomic analyses of several immune cells at different times post-infection (pi). Four sheep were infected with BTV serotype 8 and blood samples collected at days 0, 3, 7 and 15pi to perform transcriptomic analysis of B-cell marker+, CD4+, CD8+, and CD14+ sorted peripheral mononuclear cells. The maximum number of differentially expressed genes occurred at day 7pi, which coincided with the peak of infection. KEGG pathway enrichment analysis indicated that genes belonging to virus sensing and immune response initiation pathways were enriched at day 3 and 7 pi in all 4 cell population analyzed. Transcriptomic analysis also showed that at day 7pi T cell exhaustion pathway was enriched in CD4+ cells, while CD8+ cells downregulated immune response initiation pathways. T cell functional studies demonstrated that BTV produced an acute inhibition of CD4+ and CD8+ T cell activation at the peak of replication. This coincided with PD-L1 upregulation on the surface of CD4+ and CD8+ T cells as well as monocytes. Taken together, these data indicate that BTV could exploit the PD1/PD-L1 immune checkpoint to impair T cell responses. These findings identify several mechanisms in the interaction between host and BTV, which could help develop better tools to combat the disease.
- Published
- 2023
- Full Text
- View/download PDF
3. Hemagglutinin protein of Peste des Petits Ruminants virus (PPRV) activates the innate immune response via Toll-like receptor 2 signaling
- Author
-
José M. Rojas, Elena Pascual, Sean R. Wattegedera, Miguel Avia, César Santiago, Verónica Martín, Gary Entrican, and Noemí Sevilla
- Subjects
tlr2 ,morbillivirus ,hemagglutinin ,interleukin-8 ,dendritic cells ,monocyte ,macrophage ,erk ,Infectious and parasitic diseases ,RC109-216 - Abstract
The toll-like receptor (TLR) family comprises both cell-surface and intracellular receptors that recognize different types of pathogen-associated molecular patterns (PAMPs) leading to the production of pro-inflammatory cytokines and subsequent development of adaptive immunity. TLR2 is a cell-surface receptor initially thought to act as a bacterial sentinel but also shown to recognize a number of viral glycoproteins. In this study, we sought to characterize the role of TLR2 in the activation of the immune response by peste des petits ruminants virus (PPRV), a morbillivirus of the Paramixoviridae family that causes an acute, highly contagious disease in goats and sheep. Using human embryonic kidney (HEK) 293 cells stably expressing human (h)TLR2 but lacking any other TLR, we found that PPRV induces IL-8 production in a dose-dependent manner. That activation is only observed in cells expressing hTLR2 and is greatly reduced when the receptor is blocked by pretreatment with specific antibody. We identified hemagglutinin (H) as the viral protein responsible of TLR2 activation by performing the same assays with purified recombinant mammalian-expressed H protein. Exogenous addition of recombinant H protein to cell culture induces high levels of interleukin (IL)-8 only in TLR2-expressing cells. Moreover, H engagement on TLR2 in the monocytic cell line THP-1 activates extracellular-signal-regulated kinase (ERK) signaling. Stimulation of primary ovine dendritic cells with either inactivated PPRV or purified recombinant H protein results in transcription of pro-inflammatory cytokines and the secretion of the Th1-polarizing cytokine IL-12. The role of these host immune mechanisms in the control of PPR is discussed.
- Published
- 2021
- Full Text
- View/download PDF
4. Non-replicative antibiotic resistance-free DNA vaccine encoding S and N proteins induces full protection in mice against SARS-CoV-2
- Author
-
Pedro J. Alcolea, Jaime Larraga, Daniel Rodríguez-Martín, Ana Alonso, Francisco J. Loayza, José M. Rojas, Silvia Ruiz-García, Andrés Louloudes-Lázaro, Ana B. Carlón, Pedro J. Sánchez-Cordón, Pablo Nogales-Altozano, Natalia Redondo, Miguel Manzano, Daniel Lozano, Jesús Palomero, María Montoya, María Vallet-Regí, Verónica Martín, Noemí Sevilla, and Vicente Larraga
- Subjects
SARS-CoV-2 ,DNA vaccine ,S protein ,N protein ,mouse model ,pPAL ,Immunologic diseases. Allergy ,RC581-607 - Abstract
SARS-CoV-2 vaccines currently in use have contributed to controlling the COVID-19 pandemic. Notwithstanding, the high mutation rate, fundamentally in the spike glycoprotein (S), is causing the emergence of new variants. Solely utilizing this antigen is a drawback that may reduce the efficacy of these vaccines. Herein we present a DNA vaccine candidate that contains the genes encoding the S and the nucleocapsid (N) proteins implemented into the non-replicative mammalian expression plasmid vector, pPAL. This plasmid lacks antibiotic resistance genes and contains an alternative selectable marker for production. The S gene sequence was modified to avoid furin cleavage (Sfs). Potent humoral and cellular immune responses were observed in C57BL/6J mice vaccinated with pPAL-Sfs + pPAL-N following a prime/boost regimen by the intramuscular route applying in vivo electroporation. The immunogen fully protected K18-hACE2 mice against a lethal dose (105 PFU) of SARS-CoV-2. Viral replication was completely controlled in the lungs, brain, and heart of vaccinated mice. Therefore, pPAL-Sfs + pPAL-N is a promising DNA vaccine candidate for protection from COVID-19.
- Published
- 2022
- Full Text
- View/download PDF
5. Adenoviral delivery of soluble ovine OX40L or CD70 costimulatory molecules improves adaptive immune responses to a model antigen in sheep
- Author
-
José M. Rojas, Carolina Mancho, Andrés Louloudes-Lázaro, Daniel Rodríguez-Martín, Miguel Avia, Santiago Moreno, Noemí Sevilla, and Verónica Martín
- Subjects
immune response ,CD70 ,OX40L ,adenoviral vectors ,vaccine ,Microbiology ,QR1-502 - Abstract
The tumour necrosis factor superfamily OX40L and CD70 and their receptors are costimulatory signalling axes critical for adequate T and B cell activation in humans and mice. In this work we inoculated groups of sheep with human recombinant adenovirus type 5 (Ad) expressing Ovis aries (Oa)OX40L or OaCD70 or a control adenoviral vector to determine whether they could improve the immune response to the model antigen OVA. PBMCs and serum samples were obtained for analysis of the adaptive immune response to OVA at days 0, 15, 30 and 90 post-inoculation (pi). Recall responses to OVA were assessed at day 7 and 30 after the second antigen inoculation (pb) at day 90. Administration of these immunomodulatory molecules did not induce unspecific PBMC stimulation. While OaOX40L administration mainly increased TNF-α and IL-4 in PBMC at day 15 pi concomitantly with a slight increase in antibody titer and the number of IFN-γ producing cells, we detected greater effects on adaptive immunity after OaCD70 administration. AdOaCD70 inoculation improved antibody titers to OVA at days 30 and 90 pi, and increased anti-OVA-specific IgG-secreting B cell counts when compared to control. Moreover, higher IFN-γ production was detected on days 7 pi, 7 pb and 30 pb in PBMCs from this group. Phenotypic analysis of T cell activation showed an increase in effector CD8+ T cells (CD8+ CD62L- CD27-) at day 15 pi in AdOaCD70 group, concurrent with a decrease in early activated cells (CD8+ CD62L- CD27+). Moreover, recall anti-OVA CD8+ T cell responses were increased at 7 pb in the AdOaCD70 group. AdOaCD70 administration could therefore promote CD8+ T cell effector differentiation and long-term activity. In this work we characterized the in vivo adjuvant potential on the humoral and cellular immune response of OaOX40L and OaCD70 delivered by non-replicative adenovirus vectors using the model antigen OVA. We present data highlighting the potency of these molecules as veterinary vaccine adjuvant.
- Published
- 2022
- Full Text
- View/download PDF
6. Immunization With Bovine Herpesvirus-4-Based Vector Delivering PPRV-H Protein Protects Sheep From PPRV Challenge
- Author
-
Daniel Rodríguez-Martín, José Manuel Rojas, Francesca Macchi, Valentina Franceschi, Luca Russo, Noemí Sevilla, Gaetano Donofrío, and Verónica Martín
- Subjects
vaccine ,PPRV ,immune response ,ruminant ,protection correlates ,DIVA ,Immunologic diseases. Allergy ,RC581-607 - Abstract
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.
- Published
- 2021
- Full Text
- View/download PDF
7. A New Look at Vaccine Strategies Against PPRV Focused on Adenoviral Candidates
- Author
-
José M. Rojas, Noemí Sevilla, and Verónica Martín
- Subjects
PPRV ,vaccines ,adenovirus ,viral vector ,immune response ,Veterinary medicine ,SF600-1100 - Abstract
Peste des petits ruminants virus (PPRV) is a virus that mainly infects goats and sheep causing significant economic loss in Africa and Asia, but also posing a serious threat to Europe, as recent outbreaks in Georgia (2016) and Bulgaria (2018) have been reported. In order to carry out the eradication of PPRV, an objective set for 2030 by the Office International des Epizooties (OIE) and the Food and Agriculture Organization of the United Nations (FAO), close collaboration between governments, pharmaceutical companies, farmers and researchers, among others, is needed. Today, more than ever, as seen in the response to the SARS-CoV2 pandemic that we are currently experiencing, these goals are feasible. We summarize in this review the current vaccination approaches against PPRV in the field, discussing their advantages and shortfalls, as well as the development and generation of new vaccination strategies, focusing on the potential use of adenovirus as vaccine platform against PPRV and more broadly against other ruminant pathogens.
- Published
- 2021
- Full Text
- View/download PDF
8. Inhibition of the IFN Response by Bluetongue Virus: The Story So Far
- Author
-
José Manuel Rojas, Miguel Avia, Verónica Martín, and Noemí Sevilla
- Subjects
orbivirus ,BTV-NS3 ,BTV-NS4 ,double-stranded RNA ,STAT1 ,STAT2 ,Microbiology ,QR1-502 - Abstract
Bluetongue virus (BTV) is the prototypical orbivirus that belongs to the Reoviridae family. BTV infection produces a disease in ruminants, particularly in sheep, that results in economic losses through reduced productivity. BTV is transmitted by the bite of Culicoides spp. midges and is nowadays distributed globally throughout subtropical and even temperate regions. As most viruses, BTV is susceptible to the IFN response, the first line of defense employed by the immune system to combat viral infections. In turn, BTV has evolved strategies to counter the IFN response and promote its replication. The present review we will revise the works describing how BTV interferes with the IFN response.
- Published
- 2021
- Full Text
- View/download PDF
9. Vaccination With Recombinant Adenoviruses Expressing the Bluetongue Virus Subunits VP7 and VP2 Provides Protection Against Heterologous Virus Challenge
- Author
-
José Manuel Rojas, Diego Barba-Moreno, Miguel Avia, Noemí Sevilla, and Verónica Martín
- Subjects
vaccine ,Orbivirus ,T cell ,cytotoxic T lymphocytes ,IFNAR(−/−) mice ,Veterinary medicine ,SF600-1100 - Abstract
Bluetongue virus (BTV) is the causative agent of a disease that affects domestic and wild ruminants and leads to critical economic losses. BTV is an arbovirus from the Reoviridae family that is typically transmitted by the bite of infected Culicoides midges. BTV possesses multiple serotypes (up to 28 have been described), and immunity to one serotype offers little cross-protection to other serotypes. The design of vaccines that provide protection across multiple serotypes is therefore highly desirable to control this disease. We previously reported that a recombinant replication-defective human adenovirus serotype 5 (Ad5) that expresses the VP7 inner core protein of BTV serotype 8 (Ad5VP7-8) induced T-cell responses and provided protection. In the present work, we evaluated as BTV vaccine the combination of Ad5VP7-8 with another recombinant Ad5 that expresses the outer core protein VP2 from BTV-1 (Ad5VP2-1). The combination of Ad5VP2-1 and Ad5VP7-8 protected against homologous BTV challenge (BTV-1 and BTV-8) and partially against heterologous BTV-4 in a murine model. Cross-reactive anti-BTV immunoglobulin G (IgG) were detected in immunized animals, but no significant titers of neutralizing antibodies were elicited. The Ad5VP7-8 immunization induced T-cell responses that recognized all three serotypes tested in this study and primed cytotoxic T lymphocytes specific for VP7. This study further confirms that targeting antigenic determinant shared by several BTV serotypes using cellular immunity could help develop multiserotype BTV vaccines.
- Published
- 2021
- Full Text
- View/download PDF
10. A virus-encoded type I interferon decoy receptor enables evasion of host immunity through cell-surface binding
- Author
-
Bruno Hernáez, Juan Manuel Alonso-Lobo, Imma Montanuy, Cornelius Fischer, Sascha Sauer, Luis Sigal, Noemí Sevilla, and Antonio Alcamí
- Subjects
Science - Abstract
Secreted cytokine decoy receptors encoded by viruses can act as potent immune evasion proteins modulating antiviral immunity. Here Hernaez et al. show that cell surface binding is required for efficient evasion of the host response by a secreted virus encoded type I IFN decoy receptor of vaccinia and ectromelia virus using an in vivo model of infection.
- Published
- 2018
- Full Text
- View/download PDF
11. Vaccination as a Strategy to Prevent Bluetongue Virus Vertical Transmission
- Author
-
José M. Rojas, Verónica Martín, and Noemí Sevilla
- Subjects
orbivirus ,bluetongue ,vaccination ,vertical transmission ,review ,Medicine - Abstract
Bluetongue virus (BTV) produces an economically important disease in ruminants of compulsory notification to the OIE. BTV is typically transmitted by the bite of Culicoides spp., however, some BTV strains can be transmitted vertically, and this is associated with fetus malformations and abortions. The viral factors associated with the virus potency to cross the placental barrier are not well defined. The potency of vertical transmission is retained and sometimes even increased in live attenuated BTV vaccine strains. Because BTV possesses a segmented genome, the possibility of reassortment of vaccination strains with wild-type virus could even favor the transmission of this phenotype. In the present review, we will describe the non-vector-based BTV infection routes and discuss the experimental vaccination strategies that offer advantages over this drawback of some live attenuated BTV vaccines.
- Published
- 2021
- Full Text
- View/download PDF
12. Vaccination with recombinant adenovirus expressing peste des petits ruminants virus-F or -H proteins elicits T cell responses to epitopes that arises during PPRV infection
- Author
-
José Manuel Rojas, Miguel Avia, Elena Pascual, Noemí Sevilla, and Verónica Martín
- Subjects
Veterinary medicine ,SF600-1100 - Abstract
Abstract Peste des petits ruminants virus (PPRV) causes an economically important disease that limits productivity in small domestic ruminants and often affects the livestock of the poorest populations in developing countries. Animals that survive PPRV develop strong cellular and humoral responses, which are probably necessary for protection. Vaccination should thus aim at mimicking these natural responses. Immunization strategies against this morbillivirus using recombinant adenoviruses expressing PPRV-F or -H proteins can protect PPRV-challenged animals and permit differentiation of infected from vaccinated animals. Little is known of the T cell repertoire these recombinant vaccines induce. In the present work, we identified several CD4+ and CD8+ T cell epitopes in sheep infected with PPRV. We also show that recombinant adenovirus vaccination induced T cell responses to the same epitopes, and led to memory T cell differentiation. T cells primed by these recombinant adenovirus vaccines expanded after PPRV challenge and probably contributed to protection. These data validate the use of recombinant adenovirus expressing PPRV genes as DIVA strategies to control this highly contagious disease.
- Published
- 2017
- Full Text
- View/download PDF
13. Recall T cell responses to bluetongue virus produce a narrowing of the T cell repertoire
- Author
-
José-Manuel Rojas, Teresa Rodríguez-Calvo, and Noemí Sevilla
- Subjects
Veterinary medicine ,SF600-1100 - Abstract
Abstract In most viral infections, recall T cell responses are critical for protection. The magnitude of these secondary responses can also affect the CD8 and CD4 epitope repertoire diversity. Bluetongue virus (BTV) infection in sheep elicits a T cell response that contributes to viremia control and could be relevant for cross-protection between BTV serotypes. Here, we characterized CD4+ and CD8+ T cell responses during primary and recall responses. During primary immune responses, both CD4+ and CD8+ T cell populations expanded by 14 days post-infection (dpi). CD4+ T cell populations showed a lower peak of expansion and prolonged contraction phase compared to CD8+ T cell populations. Recall responses to BTV challenge led to BTV-specific expansion and activation of CD8+ but not of CD4+ T cells. The evolution of the BTV-specific TCR repertoire was also characterized in response to VP7 peptide stimulation. Striking differences in repertoire development were noted over the time-course of infection. During primary responses, a broader repertoire was induced for MHC-I and MHC-II epitopes. However, during memory responses, a narrowed repertoire was activated towards a dominant motif in VP7 comprising amino acids 139–291. Monocytes were also examined, and expanded during acute infection resolution. In addition, pro-inflammatory cytokine levels increased after BTV inoculation and persisted throughout the experiment, indicative of a prolonged inflammatory state during BTV infections. These findings could have implications for vaccine design as the narrowing memory T cell repertoire induced after BTV re-infection could lead to the development of protective immunodominant TCR repertoires that differs between individual sheep.
- Published
- 2017
- Full Text
- View/download PDF
14. The Interplay between Bluetongue Virus Infections and Adaptive Immunity
- Author
-
Daniel Rodríguez-Martín, Andrés Louloudes-Lázaro, Miguel Avia, Verónica Martín, José M. Rojas, and Noemí Sevilla
- Subjects
orbivirus ,cytotoxic T-lymphocytes ,T-helper cells ,gamma-delta T-cells ,ruminants ,B-cells ,Microbiology ,QR1-502 - Abstract
Viral infections have long provided a platform to understand the workings of immunity. For instance, great strides towards defining basic immunology concepts, such as MHC restriction of antigen presentation or T-cell memory development and maintenance, have been achieved thanks to the study of lymphocytic choriomeningitis virus (LCMV) infections. These studies have also shaped our understanding of antiviral immunity, and in particular T-cell responses. In the present review, we discuss how bluetongue virus (BTV), an economically important arbovirus from the Reoviridae family that affects ruminants, affects adaptive immunity in the natural hosts. During the initial stages of infection, BTV triggers leucopenia in the hosts. The host then mounts an adaptive immune response that controls the disease. In this work, we discuss how BTV triggers CD8+ T-cell expansion and neutralizing antibody responses, yet in some individuals viremia remains detectable after these adaptive immune mechanisms are active. We present some unpublished data showing that BTV infection also affects other T cell populations such as CD4+ T-cells or γδ T-cells, as well as B-cell numbers in the periphery. This review also discusses how BTV evades these adaptive immune mechanisms so that it can be transmitted back to the arthropod host. Understanding the interaction of BTV with immunity could ultimately define the correlates of protection with immune mechanisms that would improve our knowledge of ruminant immunology.
- Published
- 2021
- Full Text
- View/download PDF
15. Activation of OX40 and CD27 Costimulatory Signalling in Sheep through Recombinant Ovine Ligands
- Author
-
José Manuel Rojas, Alí Alejo, Jose Miguel Avia, Daniel Rodríguez-Martín, Carolina Sánchez, Antonio Alcamí, Noemí Sevilla, and Verónica Martín
- Subjects
T cell costimulatory signal ,ovine immunology ,TNF receptor ,OX40 ,CD27 ,Medicine - Abstract
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.
- Published
- 2020
- Full Text
- View/download PDF
16. Peste des Petits Ruminants Virus Fusion and Hemagglutinin Proteins Trigger Antibody-Dependent Cell-Mediated Cytotoxicity in Infected Cells
- Author
-
José M. Rojas, Daniel Rodríguez-Martín, Miguel Avia, Verónica Martín, and Noemí Sevilla
- Subjects
NK cells ,ADCC ,Morbillivirus ,bluetongue virus ,BTV ,sheep ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The adaptive immune system utilizes multiple effector mechanisms to clear viral infections. Among those antibody-dependent cell-mediated cytotoxicity (ADCC) can help recognize and clear virus-infected cells. In the present work we evaluated ADCC contribution to immunity in two economically important viral diseases that affect ruminants: bluetongue and peste des petits ruminants. Immune sera obtained from sheep experimentally infected with bluetongue virus (BTV) serotype 8 or peste des petits ruminant virus (PPRV) IC'89 were used for this study. PPRV immune sera could bind to the surface of PPRV-infected ovine B cells while BTV immune sera was unable to bind to the surface of BTV-infected sheep cells but could recognize intracellular BTV antigens. BTV and PPRV immune serum ADCC potency was established using an ovine autologous cytotoxicity assay that employed an NK cell-enriched fraction as effector cells and a virus-infected B cell-enriched fraction as target cells. In this system, immune sera triggered ADCC against PPRV-infected cells, but not against BTV-infected cells. PPRV immune sera could recognize PPRV fusion and hemagglutinin proteins on the surface of transfected cells, and enhanced lysis of these cells in ADCC assays. This indicated that these viral antigens are natural ADCC targets during PPRV infection. The present work describes a novel effector immune mechanism against PPRV in the natural host that could contribute to virus clearance highlighting the importance of studying protective immune mechanisms to improve current vaccines by invoking all effector arms of immunity.
- Published
- 2019
- Full Text
- View/download PDF
17. Bovine Herpesvirus-4-Based Vector Delivering Peste des Petits Ruminants Virus Hemagglutinin ORF Induces both Neutralizing Antibodies and Cytotoxic T Cell Responses
- Author
-
Francesca Macchi, José Manuel Rojas, Andrea Elizabeth Verna, Noemí Sevilla, Valentina Franceschi, Giulia Tebaldi, Sandro Cavirani, Verónica Martín, and Gaetano Donofrio
- Subjects
BoHV-4 ,PPRV ,DIVA vaccines ,H antigen ,viral vaccines ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Peste des Petits Ruminants Virus (PPRV) is an extremely infective morbillivirus that primarily affects goats and sheep. In underdeveloped countries where livestock are the main economical resource, PPRV causes considerable economic losses. Protective live attenuated vaccines are currently available but they induce antibody responses similar to those produced in PPRV naturally infected animals. Effective vaccines able to distinguish between vaccinated and naturally infected animals are required to PPRV control and eradication programs. Hemagglutinin (H) is a highly immunogenic PPRV envelope glycoprotein displaying both hemagglutinin and neuraminidase activities, playing a crucial role in virus attachment and penetration. In this study, a recombinant Bovine Herpesvirus-4 (BoHV-4)-based vector delivering an optimized PPRV-Hemagglutinin expression cassette, BoHV-4-A-PPRV-H-ΔTK, was assessed in immunocompetent C57BL/6 mice. BoHV-4-A-PPRV-H-ΔTK-immunization elicited both cellular and humoral immune responses with specific T cell, cytotoxic T lymphocyte, and sero-neutralizing antibody against PPRV. These data suggest recombinant BoHV-4-A-PPRV-H-ΔTK as an effective vaccine candidate to protect against PPRV herd infection and potentially applicable for eradication programs.
- Published
- 2018
- Full Text
- View/download PDF
18. IL-10: A Multifunctional Cytokine in Viral Infections
- Author
-
José M. Rojas, Miguel Avia, Verónica Martín, and Noemí Sevilla
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Abstract
The anti-inflammatory master regulator IL-10 is critical to protect the host from tissue damage during acute phases of immune responses. This regulatory mechanism, central to T cell homeostasis, can be hijacked by viruses to evade immunity. IL-10 can be produced by virtually all immune cells, and it can also modulate the function of these cells. Understanding the effects of this multifunctional cytokine is therefore a complex task. In the present review we discuss the factors driving IL-10 production and the cellular sources of the cytokine during antiviral immune responses. We particularly focus on the IL-10 regulatory mechanisms that impact antiviral immune responses and how viruses can use this central regulatory pathway to evade immunity and establish chronic/latent infections.
- Published
- 2017
- Full Text
- View/download PDF
19. Protective Efficacy in Sheep of Adenovirus-Vectored Vaccines against Bluetongue Virus Is Associated with Specific T Cell Responses.
- Author
-
Verónica Martín, Elena Pascual, Miguel Avia, Lourdes Peña, Félix Valcárcel, and Noemí Sevilla
- Subjects
Medicine ,Science - Abstract
Bluetongue virus (BTV) is an economically important Orbivirus of the Reoviridae family that causes a hemorrhagic disease in ruminants. Its control has been achieved by inactivated-vaccines that have proven to protect against homologous BTV challenge although unable to induce long-term immunity. Therefore, a more efficient control strategy needs to be developed. Recombinant adenovirus vectors are lead vaccine candidates for protection of several diseases, mainly because of their potency to induce potent T cell immunity. Here we report the induction of humoral and T-cell mediated responses able to protect animals against BTV challenge by recombinant replication-defective human adenovirus serotype 5 (Ad5) expressing either VP7, VP2 or NS3 BTV proteins. First we used the IFNAR(-/-) mouse model system to establish a proof of principle, and afterwards we assayed the protective efficacy in sheep, the natural host of BTV. Mice were completely protected against BTV challenge, developing humoral and BTV-specific CD8+- and CD4+-T cell responses by vaccination with the different rAd5. Sheep vaccinated with Ad5-BTV-VP2 and Ad5-BTV-VP7 or only with Ad5-BTV-VP7 and challenged with BTV showed mild disease symptoms and reduced viremia. This partial protection was achieved in the absence of neutralizing antibodies but strong BTV-specific CD8+ T cell responses in those sheep vaccinated with Ad5-BTV-VP7. These data indicate that rAd5 is a suitable vaccine vector to induce T cell immunity during BTV vaccination and provide new data regarding the relevance of T cell responses in protection during BTV infection.
- Published
- 2015
- Full Text
- View/download PDF
20. Vaccination with recombinant adenoviruses expressing the peste des petits ruminants virus F or H proteins overcomes viral immunosuppression and induces protective immunity against PPRV challenge in sheep.
- Author
-
José M Rojas, Héctor Moreno, Félix Valcárcel, Lourdes Peña, Noemí Sevilla, and Verónica Martín
- Subjects
Medicine ,Science - Abstract
Peste des petits ruminants (PPR) is a highly contagious disease of small ruminants caused by the Morbillivirus peste des petits ruminants virus (PPRV). Two recombinant replication-defective human adenoviruses serotype 5 (Ad5) expressing either the highly immunogenic fusion protein (F) or hemagglutinin protein (H) from PPRV were used to vaccinate sheep by intramuscular inoculation. Both recombinant adenovirus vaccines elicited PPRV-specific B- and T-cell responses. Thus, neutralizing antibodies were detected in sera from immunized sheep. In addition, we detected a significant antigen specific T-cell response in vaccinated sheep against two different PPRV strains, indicating that the vaccine induced heterologous T cell responses. Importantly, no clinical signs and undetectable virus shedding were observed after virulent PPRV challenge in vaccinated sheep. These vaccines also overcame the T cell immunosuppression induced by PPRV in control animals. The results indicate that these adenovirus constructs could be a promising alternative to current vaccine strategies for the development of PPRV DIVA vaccines.
- Published
- 2014
- Full Text
- View/download PDF
21. Mutagenesis-mediated decrease of pathogenicity as a feature of the mutant spectrum of a viral population.
- Author
-
Marta Sanz-Ramos, Teresa Rodríguez-Calvo, and Noemí Sevilla
- Subjects
Medicine ,Science - Abstract
BACKGROUND: RNA virus populations are heterogeneous ensembles of closely related genomes termed quasispecies. This highly complex distribution of variants confers important properties to RNA viruses and influences their pathogenic behavior. It has been hypothesized that increased mutagenesis of viral populations, by treatment with mutagenic agents, can induce alterations in the pathogenic potential of a virus population. In this work we investigate whether mutagenized foot-and-mouth disease virus (FMDV) populations display changes in their virulence in mice. METHODOLOGY AND PRINCIPAL FINDINGS: FMDV C-S8c1 was passaged in BHK cells in the presence of the mutagenic agent ribavirin. Decline in viral titer and viral RNA progeny was observed in the first passage, fluctuating around a constant value thereafter. Hence, the specific infectivity remained stable during the passages. The viral population harvested from passage 9 (P9 R) showed decreased virulence in mice, with a lethal dose 50 (LD(50)) >10(4) PFU, as compared with LD(50) of 50 PFU of the parental population FMDV C-S8c1. This decrease in virulence was associated to a 20-fold increase in the mutation frequency of the P9 R population with respect to C-S8c1. Interestingly, individual biological clones isolated from the attenuated population P9 R were as virulent as the parental virus C-S8c1. Furthermore, a mixed population of C-S8c1 and P9 R was inoculated into mice and showed decreased virulence as compared to C-S8c1, suggesting that population P9 R is able to suppress the virulent phenotype of C-S8c1. CONCLUSION: Ribavirin-mediated mutagenesis of an FMDV population resulted in attenuation in vivo, albeit a large proportion of its biological clones displayed a highly virulent phenotype. These results, together with the suppression of C-S8c1 by mutagenized P9 R population, document a suppressive effect of mutagenized viral quasispecies in vivo, and suggest novel approaches to the treatment and prevention of viral diseases.
- Published
- 2012
- Full Text
- View/download PDF
22. New vaccine design based on defective genomes that combines features of attenuated and inactivated vaccines.
- Author
-
Teresa Rodríguez-Calvo, Samuel Ojosnegros, Marta Sanz-Ramos, Juan García-Arriaza, Cristina Escarmís, Esteban Domingo, and Noemí Sevilla
- Subjects
Medicine ,Science - Abstract
BackgroundNew vaccine designs are needed to control diseases associated with antigenically variable RNA viruses. Foot-and-mouth disease (FMD) is a highly contagious disease of livestock that inflicts severe economic losses. Although the current whole-virus chemically inactivated vaccine has proven effective, it has led to new outbreaks of FMD because of incomplete inactivation of the virus or the escape of infectious virus from vaccine production premises. We have previously shown that serial passages of FMD virus (FMDV) C-S8c1 at high multiplicity of infection in cell culture resulted in virus populations consisting of defective genomes that are infectious by complementation (termed C-S8p260).Principal findingHere we evaluate the immunogenicity of C-S8p260, first in a mouse model system to establish a proof of principle, and second, in swine, the natural host of FMDV C-S8c1. Mice were completely protected against a lethal challenge with FMDV C-S8c1, after vaccination with a single dose of C-S8p260. Pigs immunized with different C-S8p260 doses and challenged with FMDV C-S8c1 either did not develop any clinical signs or showed delayed and mild disease symptoms. C-S8p260 induced high titers of both FMDV-specific, neutralizing antibodies and activated FMDV-specific T cells in swine, that correlated with solid protection against FMDV.ConclusionsThe defective virus-based vaccine did not produce detectable levels of transmissible FMDV. Therefore, a segmented, replication-competent form of a virus, such as FMDV C-S8p260, can provide the basis of a new generation of attenuated antiviral vaccines with two safety barriers. The design can be extended to any viral pathogen that encodes trans-acting gene products, allowing complementation between replication-competent, defective forms.
- Published
- 2010
- Full Text
- View/download PDF
23. Immunosuppression during acute infection with foot-and-mouth disease virus in swine is mediated by IL-10.
- Author
-
Fayna Díaz-San Segundo, Teresa Rodríguez-Calvo, Ana de Avila, and Noemí Sevilla
- Subjects
Medicine ,Science - Abstract
Foot-and-mouth disease virus (FMDV) is one of the most contagious animal viruses, causing a devastating disease in cloven-hoofed animals with enormous economic consequences. Identification of the different parameters involved in the immune response elicited against FMDV remains unclear, and it is fundamental the understanding of such parameters before effective control measures can be put in place. In the present study, we show that interleukin-10 (IL-10) production by dendritic cells (DCs) is drastically increased during acute infection with FMDV in swine. In vitro blockade of IL-10 with a neutralizing antibody against porcine IL-10 restores T cell activation by DCs. Additionally, we describe that FMDV infects DC precursors and interferes with DC maturation and antigen presentation capacity. Thus, we propose a new mechanism of virus immunity in which a non-persistent virus, FMDV, induces immunosuppression by an increment in the production of IL-10, which in turn, reduces T cell function. This reduction of T cell activity may result in a more potent induction of neutralizing antibody responses, clearing the viral infection.
- Published
- 2009
- Full Text
- View/download PDF
24. Establishment of a bluetongue virus infection model in mice that are deficient in the alpha/beta interferon receptor.
- Author
-
Eva Calvo-Pinilla, Teresa Rodríguez-Calvo, Juan Anguita, Noemí Sevilla, and Javier Ortego
- Subjects
Medicine ,Science - Abstract
Bluetongue (BT) is a noncontagious, insect-transmitted disease of ruminants caused by the bluetongue virus (BTV). A laboratory animal model would greatly facilitate the studies of pathogenesis, immune response and vaccination against BTV. Herein, we show that adult mice deficient in type I IFN receptor (IFNAR((-/-))) are highly susceptible to BTV-4 and BTV-8 infection when the virus is administered intravenously. Disease was characterized by ocular discharges and apathy, starting at 48 hours post-infection and quickly leading to animal death within 60 hours of inoculation. Infectious virus was recovered from the spleen, lung, thymus, and lymph nodes indicating a systemic infection. In addition, a lymphoid depletion in spleen, and severe pneumonia were observed in the infected mice. Furthermore, IFNAR((-/-)) adult mice immunized with a BTV-4 inactivated vaccine showed the induction of neutralizing antibodies against BTV-4 and complete protection against challenge with a lethal dose of this virus. The data indicate that this mouse model may facilitate the study of BTV pathogenesis, and the development of new effective vaccines for BTV.
- Published
- 2009
- Full Text
- View/download PDF
25. A Morbillivirus Infection Shifts DC Maturation Toward a Tolerogenic Phenotype to Suppress T Cell Activation
- Author
-
Daniel Rodríguez-Martín, Isabel García-García, Verónica Martín, José Manuel Rojas, and Noemí Sevilla
- Subjects
Immunosuppression Therapy ,Sheep ,Goats ,T-Lymphocytes ,Immunology ,Cell Differentiation ,Dendritic Cells ,Monocyte ,Lymphocyte Activation ,Monocyte-derived DC ,Antiviral Agents ,Microbiology ,Peste-des-petits-ruminants virus ,Phenotype ,Virology ,Insect Science ,Peste-des-Petits-Ruminants ,Concanavalin A ,Pathogenesis and Immunity ,Animals ,PPRV ,Monocyte-derived dendritic cell ,Mitogens ,Tolerance ,Immunosuppression - Abstract
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.
- Published
- 2022
- Full Text
- View/download PDF
26. Hemagglutinin protein of Peste des Petits Ruminants virus (PPRV) activates the innate immune response via Toll-like receptor 2 signaling
- Author
-
Elena Pascual, Sean Wattegedera, Noemí Sevilla, Gary Entrican, José M. Rojas, Miguel Avia, César Santiago, Verónica Martín, Ministerio de Ciencia y Tecnología (España), European Commission, Comunidad de Madrid, Wattegedera, Sean R [0000-0002-2218-5144], Avia, Miguel [0000-0001-7661-4779], Entrican, Gary [0000-0002-8822-2331], Sevilla, Noemí [0000-0003-0118-0376], Wattegedera, Sean R, Avia, Miguel, Entrican, Gary, and Sevilla, Noemí
- Subjects
THP-1 Cells ,Macrophage ,Infectious and parasitic diseases ,RC109-216 ,Interleukin 8 ,Monocyte ,Dendritic cells ,TLR2 ,Hemagglutinin ,0303 health sciences ,Toll-like receptor ,Acquired immune system ,ERK ,Infectious Diseases ,monocyte ,Cytokines ,Signal Transduction ,Research Article ,Research Paper ,Microbiology (medical) ,Immunology ,Hemagglutinins, Viral ,Hemagglutinin (influenza) ,tlr2 ,macrophage ,Biology ,Microbiology ,Peste-des-petits-ruminants virus ,erk ,03 medical and health sciences ,Immune system ,Animals ,Humans ,hemagglutinin ,dendritic cells ,030304 developmental biology ,Sheep ,Innate immune system ,030306 microbiology ,Interleukin-8 ,HEK 293 cells ,interleukin-8 ,Virology ,Immunity, Innate ,Toll-Like Receptor 2 ,morbillivirus ,HEK293 Cells ,Morbillivirus ,Cell culture ,biology.protein ,Parasitology - Abstract
Centro de Investigación en Sanidad Animal (CISA), The toll-like receptor (TLR) family comprises both cell-surface and intracellular receptors that recognize different types of pathogen-associated molecular patterns (PAMPs) leading to the production of pro-inflammatory cytokines and subsequent development of adaptive immunity. TLR2 is a cell-surface receptor initially thought to act as a bacterial sentinel but also shown to recognize a number of viral glycoproteins. In this study, we sought to characterize the role of TLR2 in the activation of the immune response by peste des petits ruminants virus (PPRV), a morbillivirus of the Paramixoviridae family that causes an acute, highly contagious disease in goats and sheep. Using human embryonic kidney (HEK) 293 cells stably expressing human (h)TLR2 but lacking any other TLR, we found that PPRV induces IL-8 production in a dose-dependent manner. That activation is only observed in cells expressing hTLR2 and is greatly reduced when the receptor is blocked by pretreatment with specific antibody. We identified hemagglutinin (H) as the viral protein responsible of TLR2 activation by performing the same assays with purified recombinant mammalian-expressed H protein. Exogenous addition of recombinant H protein to cell culture induces high levels of interleukin (IL)-8 only in TLR2-expressing cells. Moreover, H engagement on TLR2 in the monocytic cell line THP-1 activates extracellular-signal-regulated kinase (ERK) signaling. Stimulation of primary ovine dendritic cells with either inactivated PPRV or purified recombinant H protein results in transcription of pro-inflammatory cytokines and the secretion of the Th1-polarizing cytokine IL-12. The role of these host immune mechanisms in the control of PPR is discussed., This work was funded by grants AGL2015-64290R and RTI2018-094616-B-100 from the Ministerio de Ciencia (Spain); grant VetBioNet INFRAIA-731014 from the EU-H2020 and grant S2018/BAA-4370-PLATESA2 from Comunidad de Madrid (Fondo Europeo de Desarrollo Regional, FEDER). MA was funded by an FPI grant (BES-2013-066406). NS was the holder of a fellowship from OECD Co-operative Research Programme: Biological Resource Management for Sustainable Agricultural Systems., 15 Pág.
- Published
- 2021
- Full Text
- View/download PDF
27. Diagnosing bluetongue virus in domestic ruminants: current perspectives
- Author
-
Verónica Martín, Noemí Sevilla, Daniel Rodríguez-Martín, and José M. Rojas
- Subjects
Serotype ,Orbivirus ,biology ,040301 veterinary sciences ,viruses ,0402 animal and dairy science ,Outbreak ,04 agricultural and veterinary sciences ,biology.organism_classification ,medicine.disease ,040201 dairy & animal science ,Virology ,Virus ,law.invention ,0403 veterinary science ,law ,medicine ,African horse sickness ,Virus classification ,Epizootic ,Polymerase chain reaction - Abstract
This review provides an overview of current and potential new diagnostic techniques against bluetongue virus (BTV), an Orbivirus transmitted by arthropods that affects ruminants. Bluetongue is a disease currently notifiable to the World Organization for Animal Health (OIE), causing great economic losses due to decreased trade associated with bluetongue outbreaks and high mortality and morbidity. BTV cross-reacts with many antigenically related viruses including viruses that causes African Horse sickness and epizootic haemorrhagic disease of deer. Therefore, reliable diagnostic approaches to detect BTV among these other antigenically related viruses are used or being developed. The antigenic determinant for differentiation of virus species/serogroups among orbiviruses is the VP7 protein, meanwhile VP2 is serotype specific. Serologically, assays are established in many laboratories, based mainly on competitive ELISA or serum neutralization assay (virus neutralization assay [VNT]) although new techniques are being developed. Virus isolation from blood or semen is, additionally, another means of BTV diagnosis. Nevertheless, most of these techniques for viral isolation are time-consuming and expensive. Currently, reverse-transcription polymerase chain reaction (RT-PCR) panels or real-time RT-PCR are widely used methods although next-generation sequencing remains of interest for future virus diagnosis.
- Published
- 2019
- Full Text
- View/download PDF
28. The Interplay between Bluetongue Virus Infections and Adaptive Immunity
- Author
-
Noemí Sevilla, Daniel Rodríguez-Martín, Miguel Avia, Verónica Martín, Andrés Louloudes-Lázaro, José M. Rojas, Ministerio de Ciencia e Innovación (España), Comunidad de Madrid, European Commission, Louloudes-Lázaro, Andrés, Avia, Miguel, Martín, Verónica, Rojas, José M., and Sevilla, Noemí
- Subjects
0301 basic medicine ,040301 veterinary sciences ,T-Lymphocytes ,Antigen presentation ,B-cells ,Viremia ,chemical and pharmacologic phenomena ,Review ,Adaptive Immunity ,Lymphocytic choriomeningitis ,Antibodies, Viral ,T-helper cells ,Arbovirus ,Microbiology ,Bluetongue ,Virus ,orbivirus ,0403 veterinary science ,03 medical and health sciences ,Immunity ,Virology ,cytotoxic T-lymphocytes ,medicine ,Animals ,Neutralizing antibody ,Antigen Presentation ,Sheep ,biology ,T-cells ,04 agricultural and veterinary sciences ,Ruminants ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,Acquired immune system ,Antibodies, Neutralizing ,QR1-502 ,030104 developmental biology ,Infectious Diseases ,gamma-delta T-cells ,biology.protein ,Bluetongue virus - Abstract
17 Pág. Centro de Investigación en Sanidad Animal (CISA), Viral infections have long provided a platform to understand the workings of immunity. For instance, great strides towards defining basic immunology concepts, such as MHC restriction of antigen presentation or T-cell memory development and maintenance, have been achieved thanks to the study of lymphocytic choriomeningitis virus (LCMV) infections. These studies have also shaped our understanding of antiviral immunity, and in particular T-cell responses. In the present review, we discuss how bluetongue virus (BTV), an economically important arbovirus from the Reoviridae family that affects ruminants, affects adaptive immunity in the natural hosts. During the initial stages of infection, BTV triggers leucopenia in the hosts. The host then mounts an adaptive immune response that controls the disease. In this work, we discuss how BTV triggers CD8+ T-cell expansion and neutralizing antibody responses, yet in some individuals viremia remains detectable after these adaptive immune mechanisms are active. We present some unpublished data showing that BTV infection also affects other T cell populations such as CD4+ T-cells or γδ T-cells, as well as B-cell numbers in the periphery. This review also discusses how BTV evades these adaptive immune mechanisms so that it can be transmitted back to the arthropod host. Understanding the interaction of BTV with immunity could ultimately define the correlates of protection with immune mechanisms that would improve our knowledge of ruminant immunology., This work was funded by grants RTI2018-094616-B-100 from the Ministerio de Ciencia (Spain) and S2018/BAA-4370-PLATESA2 from Comunidad de Madrid (Fondo Europeo de Desarrollo Regional, FEDER). MA was funded by an FPI grant (BES-2013-066406). ALL was funded by an FPI grant (INIA).
- Published
- 2021
29. Immunization With Bovine Herpesvirus-4-Based Vector Delivering PPRV-H Protein Protects Sheep From PPRV Challenge
- Author
-
Valentina Franceschi, Luca Russo, Noemí Sevilla, José M. Rojas, Gaetano Donofrio, Verónica Martín, Francesca Macchi, Daniel Rodríguez-Martín, European Commission, Ministerio de Ciencia e Innovación (España), Comunidad de Madrid, Università degli Studi di Parma, Rodríguez-Martín, Daniel, Rojas, José Manuel, Macchi, Francesca, Franceschi, Valentina, Russo, Luca, Sevilla, Noemí, and Donofrío, Gaetano
- Subjects
Genetic Vectors ,Immunology ,Hemagglutinin (influenza) ,Sheep Diseases ,ruminant ,DIVA ,immune response ,Peste-des-petits-ruminants virus ,Viral Proteins ,Dogs ,Morbillivirus ,vaccine ,Chlorocebus aethiops ,Peste-des-Petits-Ruminants ,Immunology and Allergy ,Animals ,Vector (molecular biology) ,Viral shedding ,Vero Cells ,Original Research ,Sheep ,biology ,Viral Vaccines ,RC581-607 ,biology.organism_classification ,Virology ,recombinant herpesvirus ,Herpesvirus 4, Bovine ,Vaccination ,Diva ,protection correlates ,Immunization ,biology.protein ,PPRV ,Immunologic diseases. Allergy - Abstract
14 Pág. Centro de Investigación en Sanidad Animal (CISA), 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., This work was funded mainly by a TNA (Translational Access Activities) from VetBioNet INFRAIA-731014 from the EU-H2020. The study also received the following grants and funding: AGL2015-64290R and RTI2018-094616-B-100 from the Ministerio de Ciencia (Spain), grant S2018/BAA-4370-PLATESA2 from Comunidad de Madrid (Fondo Europeo de Desarrollo Regional, FEDER), and internal funding from Parma University.
- Published
- 2021
30. Vaccination With Recombinant Adenoviruses Expressing the Bluetongue Virus Subunits VP7 and VP2 Provides Protection Against Heterologous Virus Challenge
- Author
-
José Manuel Rojas, Diego Barba-Moreno, Miguel Avia, Noemí Sevilla and Verónica Martín
- Abstract
Bluetongue virus (BTV) is the causative agent of a disease that affects domestic and wild ruminants and leads to critical economic losses. BTV is an arbovirus from the Reoviridae family that is typically transmitted by the bite of infectedCulicoidesmidges. BTV possesses multiple serotypes (up to 28 have been described), and immunity to one serotype offers little cross-protection to other serotypes. The design of vaccines that provide protection across multiple serotypes is therefore highly desirable to control this disease. We previously reported that a recombinant replication-defective human adenovirus serotype 5 (Ad5) that expresses the VP7 inner core protein of BTV serotype 8 (Ad5VP7-8) induced T-cell responses and provided protection. In the present work, we evaluated as BTV vaccine the combination of Ad5VP7-8 with another recombinant Ad5 that expresses the outer core protein VP2 from BTV-1 (Ad5VP2-1). The combination of Ad5VP2-1 and Ad5VP7-8 protected against homologous BTV challenge (BTV-1 and BTV-8) and partially against heterologous BTV-4 in a murine model. Cross-reactive anti-BTV immunoglobulin G (IgG) were detected in immunized animals, but no significant titers of neutralizing antibodies were elicited. The Ad5VP7-8 immunization induced T-cell responses that recognized all three serotypes tested in this study and primed cytotoxic T lymphocytes specific for VP7. This study further confirms that targeting antigenic determinant shared by several BTV serotypes using cellular immunity could help develop multiserotype BTV vaccines.  
- Published
- 2021
31. Vaccination With Recombinant Adenoviruses Expressing the Bluetongue Virus Subunits VP7 and VP2 Provides Protection Against Heterologous Virus Challenge
- Author
-
Diego Barba-Moreno, José M. Rojas, Miguel Avia, Verónica Martín, Noemí Sevilla, Ministerio de Ciencia e Innovación (España), European Commission, Comunidad de Madrid, Ministerio de Economía y Competitividad (España), Rojas, José Manuel [0000-0002-4055-3967], Avia, Miguel [0000-0001-7661-4779], Sevilla, Noemí [0000-0003-0118-0376], Rojas, José Manuel, Avia, Miguel, and Sevilla, Noemí
- Subjects
Serotype ,Cellular immunity ,Heterologous ,Reoviridae ,cytotoxic T lymphocytes ,Arbovirus ,Virus ,03 medical and health sciences ,IFNAR(−/−) mice ,vaccine ,medicine ,Orbivirus ,030304 developmental biology ,Original Research ,0303 health sciences ,lcsh:Veterinary medicine ,biology ,General Veterinary ,030306 microbiology ,T cell ,biology.organism_classification ,medicine.disease ,Virology ,3. Good health ,Vaccination ,Cytotoxic T lymphocytes ,IFNAR (−/−) mice ,lcsh:SF600-1100 ,Veterinary Science ,Vaccine - Abstract
Centro de Investigación en Sanidad Animal (CISA), Bluetongue virus (BTV) is the causative agent of a disease that affects domestic and wild ruminants and leads to critical economic losses. BTV is an arbovirus from the Reoviridae family that is typically transmitted by the bite of infected Culicoides midges. BTV possesses multiple serotypes (up to 28 have been described), and immunity to one serotype offers little cross-protection to other serotypes. The design of vaccines that provide protection across multiple serotypes is therefore highly desirable to control this disease. We previously reported that a recombinant replication-defective human adenovirus serotype 5 (Ad5) that expresses the VP7 inner core protein of BTV serotype 8 (Ad5VP7-8) induced T-cell responses and provided protection. In the present work, we evaluated as BTV vaccine the combination of Ad5VP7-8 with another recombinant Ad5 that expresses the outer core protein VP2 from BTV-1 (Ad5VP2-1). The combination of Ad5VP2-1 and Ad5VP7-8 protected against homologous BTV challenge (BTV-1 and BTV-8) and partially against heterologous BTV-4 in a murine model. Cross-reactive anti-BTV immunoglobulin G (IgG) were detected in immunized animals, but no significant titers of neutralizing antibodies were elicited. The Ad5VP7-8 immunization induced T-cell responses that recognized all three serotypes tested in this study and primed cytotoxic T lymphocytes specific for VP7. This study further confirms that targeting antigenic determinant shared by several BTV serotypes using cellular immunity could help develop multiserotype BTV vaccines., This work was funded by grants AGL2015-64290R and from the Ministerio de Ciencia (Spain), grant VetBioNet INFRAIA-731014 from the EU-H2020, and grant S2018/BAA-4370-PLATESA2 from Comunidad de Madrid (Fondo Europeo de Desarrollo Regional, FEDER). MA was funded by an FPI grant (BES-2013-066406)., 15 Pág.
- Published
- 2020
32. Viral pathogen-induced mechanisms to antagonize mammalian interferon (IFN) signaling pathway
- Author
-
Noemí Sevilla, Verónica Martín, Alí Alejo, José M. Rojas, Ministerio de Ciencia e Innovación (España), Comunidad de Madrid, European Commission, Rojas, José M., Alejo, Alí, and Sevilla, Noemí
- Subjects
Cell ,Review ,Biology ,Virus ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Immune system ,Interferon ,medicine ,Humans ,Receptor ,Molecular Biology ,Pathogen ,Immune Evasion ,Pharmacology ,0303 health sciences ,030302 biochemistry & molecular biology ,Antiviral responses ,Interferon-alpha ,Biological activity ,Cell Biology ,IFN-I pathway ,Immunity, Innate ,3. Good health ,Cell biology ,medicine.anatomical_structure ,Virus Diseases ,Viral evasion mechanisms ,Host-Pathogen Interactions ,Molecular Medicine ,Interferons ,Signal transduction ,medicine.drug ,Signal Transduction - Abstract
22 Pág. Centro de Investigación en Sanidad Animal (CISA), 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-β), 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., This work was supported by grant RTI2018-094616-B-100 from the Spanish Ministerio de Ciencia e Innovación; grant S2018/BAA-4370-PLATESA2 from the Comunidad de Madrid (Fondo Europeo de Desarrollo Regional, FEDER) and VetBionet INFRAIA-731014 from the European Union H2020.
- Published
- 2020
33. Activation of OX40 and CD27 Costimulatory Signalling in Sheep through Recombinant Ovine Ligands
- Author
-
Daniel Rodríguez-Martín, José M. Rojas, Alí Alejo, Jose Miguel Avia, Verónica Martín, Antonio Alcami, Carolina Sánchez, Noemí Sevilla, Ministerio de Economía y Competitividad (España), Ministerio de Ciencia, Innovación y Universidades (España), European Commission, Comunidad de Madrid, and Agencia Estatal de Investigación (España)
- Subjects
0301 basic medicine ,T cell ,Immunology ,lcsh:Medicine ,Article ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,law ,Drug Discovery ,medicine ,Pharmacology (medical) ,OX40 ,Receptor ,CD27 ,Ovine immunology ,Pharmacology ,Chemistry ,TNF receptor ,lcsh:R ,Transfection ,Hedgehog signaling pathway ,Cell biology ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Recombinant DNA ,T cell costimulatory signal ,Tumor necrosis factor alpha ,ovine immunology ,CD8 - Abstract
© 2020 by the authors., 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., M.A. was funded by an FPI Grant (BES-2013-066406) from Spanish Ministerio de Economía y Competitividad. This work was funded by grants AGL2015-64290R, RTI2018-094616-B-I00, ADEDONET-Redes de Excelencia from the Spanish Ministerio de Economía y Competitividad; grant S2013/ABI-2906-PLATESA and S2018/BAA-4370-PLATESA2 from the Comunidad de Madrid and VetBionet INFRAIA-731014 from the European Union (Fondo Europeo de Desarrollo Regional, FEDER).
- Published
- 2020
- Full Text
- View/download PDF
34. Virus-induced autophagic degradation of STAT2 as a mechanism for interferon signaling blockade
- Author
-
Verónica Martín, Miguel Avia, Adolfo García-Sastre, José M. Rojas, Elena Pascual, Noemí Sevilla, Piet A. van Rijn, Lisa Miorin, and 24551287 - Van Rijn, Petrus Antonius
- Subjects
viruses ,Biology ,Biochemistry ,Models, Biological ,Virus ,orbivirus ,03 medical and health sciences ,Viral Proteins ,0302 clinical medicine ,STAT2 ,Interferon ,Genetics ,medicine ,Autophagy ,Animals ,Humans ,STAT1 ,Phosphorylation ,Molecular Biology ,Orbivirus ,030304 developmental biology ,0303 health sciences ,NS3 ,Ubiquitination ,STAT2 Transcription Factor ,Interferon-beta ,Articles ,biology.organism_classification ,Lysosome ,Cell biology ,Virology & Molecular Biology ,Virologie & Moleculaire Biologie ,Virus Diseases ,IFN‐I ,Host-Pathogen Interactions ,Proteolysis ,biology.protein ,IFN-I ,lysosome ,Interferons ,Signal transduction ,Lysosomes ,030217 neurology & neurosurgery ,Bluetongue virus ,medicine.drug ,Signal Transduction - Abstract
The mammalian interferon (IFN) signaling pathway is a primary component of the innate antiviral response, and viral pathogens have evolved multiple mechanisms to antagonize this pathway and to facilitate infection. Bluetongue virus (BTV), an orbivirus of the Reoviridae family, is transmitted by midges to ruminants and causes a disease that produces important economic losses and restriction to animal trade and is of compulsory notification to the World Organization for Animal Health (OIE). Here, we show that BTV interferes with IFN-I and IFN-II responses in two ways, by blocking STAT1 phosphorylation and by degrading STAT2. BTV-NS3 protein, which is involved in virion egress, interacts with STAT2, and induces its degradation by an autophagy-dependent mechanism. This STAT2 degradative process requires the recruitment of an E3-Ub-ligase to NS3 as well as NS3 K63 polyubiquitination. Taken together, our study identifies a new mechanism by which a virus degrades STAT2 for IFN signaling blockade, highlighting the diversity of mechanisms employed by viruses to subvert the IFN response.
- Published
- 2019
35. Adenovirus as Tools in Animal Health
- Author
-
Noemí Sevilla, Verónica Martín, and José M. Rojas
- Subjects
0303 health sciences ,03 medical and health sciences ,medicine.medical_specialty ,Animal health ,030306 microbiology ,business.industry ,InformationSystems_INFORMATIONSTORAGEANDRETRIEVAL ,Medicine ,business ,Intensive care medicine ,GeneralLiterature_REFERENCE(e.g.,dictionaries,encyclopedias,glossaries) ,030304 developmental biology - Published
- 2019
- Full Text
- View/download PDF
36. IL-10: A Multifunctional Cytokine in Viral Infections
- Author
-
Miguel Avia, Verónica Martín, José M. Rojas, and Noemí Sevilla
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,T-Lymphocytes ,animal diseases ,medicine.medical_treatment ,Immunology ,chemical and pharmacologic phenomena ,Review Article ,Biology ,Immunomodulation ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Immunity ,medicine ,Animals ,Homeostasis ,Humans ,Immunology and Allergy ,Immune Evasion ,Mechanism (biology) ,food and beverages ,General Medicine ,biochemical phenomena, metabolism, and nutrition ,Interleukin-10 ,Virus Latency ,Cell biology ,Interleukin 10 ,030104 developmental biology ,Cytokine ,Virus Diseases ,bacteria ,Regulatory Pathway ,lcsh:RC581-607 ,Function (biology) ,030215 immunology - Abstract
The anti-inflammatory master regulator IL-10 is critical to protect the host from tissue damage during acute phases of immune responses. This regulatory mechanism, central to T cell homeostasis, can be hijacked by viruses to evade immunity. IL-10 can be produced by virtually all immune cells, and it can also modulate the function of these cells. Understanding the effects of this multifunctional cytokine is therefore a complex task. In the present review we discuss the factors driving IL-10 production and the cellular sources of the cytokine during antiviral immune responses. We particularly focus on the IL-10 regulatory mechanisms that impact antiviral immune responses and how viruses can use this central regulatory pathway to evade immunity and establish chronic/latent infections.
- Published
- 2017
- Full Text
- View/download PDF
37. A virus-encoded type I interferon decoy receptor enables evasion of host immunity through cell-surface binding
- Author
-
Antonio Alcami, Juan Manuel Alonso-Lobo, Sascha Sauer, Imma Montanuy, Cornelius Fischer, Bruno Hernáez, Noemí Sevilla, Luis J. Sigal, European Commission, and Ministerio de Economía y Competitividad (España)
- Subjects
0301 basic medicine ,Science ,animal diseases ,viruses ,medicine.medical_treatment ,Cell ,General Physics and Astronomy ,Virus Attachment ,Poxviridae Infections ,General Biochemistry, Genetics and Molecular Biology ,Type I interferon binding ,Virus ,Article ,03 medical and health sciences ,Mice ,Viral Proteins ,Immune system ,Interferon ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Decoy receptors ,lcsh:Science ,Receptor ,Vero Cells ,Glycosaminoglycans ,Mice, Inbred BALB C ,Multidisciplinary ,Chemistry ,Poxviridae ,General Chemistry ,3. Good health ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Cytokine ,Interferon Type I ,lcsh:Q ,Female ,Technology Platforms ,medicine.drug ,HeLa Cells - Abstract
Soluble cytokine decoy receptors are potent immune modulatory reagents with therapeutic applications. Some virus-encoded secreted cytokine receptors interact with glycosaminoglycans expressed at the cell surface, but the biological significance of this activity in vivo is poorly understood. Here, we show the type I interferon binding protein (IFNα/βBP) encoded by vaccinia and ectromelia viruses requires of this cell binding activity to confer full virulence to these viruses and to retain immunomodulatory activity. Expression of a variant form of the IFNα/βBP that inhibits IFN activity, but does not interact with cell surface glycosaminoglycans, results in highly attenuated viruses with a virulence similar to that of the IFNα/βBP deletion mutant viruses. Transcriptomics analysis and infection of IFN receptor-deficient mice confirmed that the control of IFN activity is the main function of the IFNα/βBP in vivo. We propose that retention of secreted cytokine receptors at the cell surface may largely enhance their immunomodulatory activity., Ministry of Economy and Competitiveness and European Union (European Regional Development’s Funds, FEDER) (grant SAF2015-67485-R) and by European Sequencing and Genotyping Infrastructure
- Published
- 2018
38. Peste des Petits Ruminants Virus Fusion and Hemagglutinin Proteins Trigger Antibody-Dependent Cell-Mediated Cytotoxicity in Infected Cells
- Author
-
Verónica Martín, Noemí Sevilla, Miguel Avia, Daniel Rodríguez-Martín, and José M. Rojas
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,Serotype ,sheep ,Immunology ,Hemagglutinin (influenza) ,Hemagglutinins, Viral ,Sheep Diseases ,chemical and pharmacologic phenomena ,NK cells ,Antibodies, Viral ,Virus ,Cell Line ,Immunophenotyping ,Peste-des-petits-ruminants virus ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,bluetongue virus ,Antigen ,Immunity ,Peste-des-Petits-Ruminants ,Immunology and Allergy ,Animals ,Humans ,Antigens, Viral ,Original Research ,Antibody-dependent cell-mediated cytotoxicity ,biology ,Immune Sera ,Antibody-Dependent Cell Cytotoxicity ,Acquired immune system ,Virology ,Killer Cells, Natural ,030104 developmental biology ,Morbillivirus ,Immunoglobulin G ,biology.protein ,PPRV ,lcsh:RC581-607 ,ADCC ,Biomarkers ,030215 immunology ,BTV - Abstract
The adaptive immune system utilizes multiple effector mechanisms to clear viral infections. Among those antibody-dependent cell-mediated cytotoxicity (ADCC) can help recognize and clear virus-infected cells. In the present work we evaluated ADCC contribution to immunity in two economically important viral diseases that affect ruminants: bluetongue and peste des petits ruminants. Immune sera obtained from sheep experimentally infected with bluetongue virus (BTV) serotype 8 or peste des petits ruminant virus (PPRV) IC'89 were used for this study. PPRV immune sera could bind to the surface of PPRV-infected ovine B cells while BTV immune sera was unable to bind to the surface of BTV-infected sheep cells but could recognize intracellular BTV antigens. BTV and PPRV immune serum ADCC potency was established using an ovine autologous cytotoxicity assay that employed an NK cell-enriched fraction as effector cells and a virus-infected B cell-enriched fraction as target cells. In this system, immune sera triggered ADCC against PPRV-infected cells, but not against BTV-infected cells. PPRV immune sera could recognize PPRV fusion and hemagglutinin proteins on the surface of transfected cells, and enhanced lysis of these cells in ADCC assays. This indicated that these viral antigens are natural ADCC targets during PPRV infection. The present work describes a novel effector immune mechanism against PPRV in the natural host that could contribute to virus clearance highlighting the importance of studying protective immune mechanisms to improve current vaccines by invoking all effector arms of immunity.
- Published
- 2018
39. PPRV-Induced Immunosuppression at the Interface of Virus-Host Interaction
- Author
-
Noemí Sevilla, Verónica Martín, and José M. Rojas
- Subjects
Interface (Java) ,medicine.medical_treatment ,medicine ,General Materials Science ,Immunosuppression ,Biology ,Virus-host interaction ,Virology - Published
- 2016
- Full Text
- View/download PDF
40. Pathogenesis of New World Arenaviruses in Humans
- Author
-
Esteban Domingo, Noemí Sevilla, Stefan Kunz, and Verónica Martín
- Subjects
Pathogenesis ,Lethal mutagenesis ,New World Arenavirus ,Humoral immunity ,Immunology ,Cellular receptor ,Biology ,Virology - Published
- 2015
- Full Text
- View/download PDF
41. Vaccination with recombinant adenovirus expressing peste des petits ruminants virus-F or -H proteins elicits T cell responses to epitopes that arises during PPRV infection
- Author
-
Noemí Sevilla, José M. Rojas, Verónica Martín, Miguel Avia, Elena Pascual, Centro de Investigacion en Sanidad Animal (INIA-CISA), and Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria = National Institute for Agricultural and Food Research and Technology (INIA)
- Subjects
0301 basic medicine ,T-Lymphocytes ,[SDV]Life Sciences [q-bio] ,T cell ,Epitopes, T-Lymphocyte ,Sheep Diseases ,Immunity, Heterologous ,Epitope ,Peste-des-petits-ruminants virus ,law.invention ,Mice ,03 medical and health sciences ,Morbillivirus ,law ,Peste-des-Petits-Ruminants ,medicine ,Animals ,Vaccines, Synthetic ,Sheep ,lcsh:Veterinary medicine ,General Veterinary ,biology ,Viral Vaccines ,biology.organism_classification ,Virology ,3. Good health ,Vaccination ,030104 developmental biology ,medicine.anatomical_structure ,Immunization ,Recombinant DNA ,lcsh:SF600-1100 ,CD8 ,Research Article - Abstract
Peste des petits ruminants virus (PPRV) causes an economically important disease that limits productivity in small domestic ruminants and often affects the livestock of the poorest populations in developing countries. Animals that survive PPRV develop strong cellular and humoral responses, which are probably necessary for protection. Vaccination should thus aim at mimicking these natural responses. Immunization strategies against this morbillivirus using recombinant adenoviruses expressing PPRV-F or -H proteins can protect PPRV-challenged animals and permit differentiation of infected from vaccinated animals. Little is known of the T cell repertoire these recombinant vaccines induce. In the present work, we identified several CD4+ and CD8+ T cell epitopes in sheep infected with PPRV. We also show that recombinant adenovirus vaccination induced T cell responses to the same epitopes, and led to memory T cell differentiation. T cells primed by these recombinant adenovirus vaccines expanded after PPRV challenge and probably contributed to protection. These data validate the use of recombinant adenovirus expressing PPRV genes as DIVA strategies to control this highly contagious disease. Electronic supplementary material The online version of this article (10.1186/s13567-017-0482-x) contains supplementary material, which is available to authorized users.
- Published
- 2017
- Full Text
- View/download PDF
42. Follicular dendritic cell disruption as a novel mechanism of virus-induced immunosuppression
- Author
-
Noemí Sevilla, Marco Caporale, Giuseppe Marruchella, Eleonora Melzi, Verónica Martín, Andrea Di Provvido, Massimo Palmarini, Virginia Gamino, Mara Rocchi, and Gary Entrican
- Subjects
0301 basic medicine ,Gene Expression Regulation, Viral ,medicine.medical_treatment ,viruses ,Biology ,Bluetongue ,Virus ,Animal Diseases ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Arbovirus ,Follicular dendritic cells ,Immunosuppression ,Lymph node ,Multidisciplinary ,Antigen ,medicine ,Immune Tolerance ,Animals ,Viremia ,B-Lymphocytes ,Sheep ,Virulence ,Macrophages ,Germinal center ,Endothelial Cells ,biochemical phenomena, metabolism, and nutrition ,Virology ,Immunohistochemistry ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,PNAS Plus ,Virus Diseases ,Immunology ,Host-Pathogen Interactions ,Viruses ,biology.protein ,Lymph Nodes ,Antibody ,Stromal Cells ,Bluetongue virus ,Dendritic Cells, Follicular ,030215 immunology - Abstract
Arboviruses cause acute diseases that increasingly affect global health. We used bluetongue virus (BTV) and its natural sheep host to reveal a previously uncharacterized mechanism used by an arbovirus to manipulate host immunity. Our study shows that BTV, similarly to other antigens delivered through the skin, is transported rapidly via the lymph to the peripheral lymph nodes. Here, BTV infects and disrupts follicular dendritic cells, hindering B-cell division in germinal centers, which results in a delayed production of high affinity and virus neutralizing antibodies. Moreover, the humoral immune response to a second antigen is also hampered in BTV-infected animals. Thus, an arbovirus can evade the host antiviral response by inducing an acute immunosuppression. Although transient, this immunosuppression occurs at the critical early stages of infection when a delayed host humoral immune response likely affects virus systemic dissemination and the clinical outcome of disease.
- Published
- 2016
43. Evidence of Activation and Suppression during the Early Immune Response to Foot-and-Mouth Disease Virus
- Author
-
T. de los Santos, Artur Summerfield, William T. Golde, L. Robinson, Noemí Sevilla, Bryan Charleston, and Marvin J. Grubman
- Subjects
Cellular immunity ,Innate immune system ,General Veterinary ,General Immunology and Microbiology ,Viral pathogenesis ,General Medicine ,Biology ,biology.organism_classification ,Virology ,Virus ,Immune system ,Viral replication ,Immunity ,Immunology ,Foot-and-mouth disease virus - Abstract
Foot-and-mouth disease virus causes a serious disease of livestock species, threatening free global trade and food security. The disease spreads rapidly between animals, and to ensure a window of opportunity for such spread, the virus has evolved multiple mechanisms to subvert the early immune response. The cycle of infection in the individual animal is very short, infection is initiated, disseminated throughout the body and infectious virus produced in
- Published
- 2011
- Full Text
- View/download PDF
44. A recombinant adenovirus expressing ovine interferon tau prevents influenza virus-induced lethality in mice
- Author
-
Verónica Martín, Elena Pascual, Giselle Rangel, A. de Molina, Noemí Sevilla, Miguel Avia, and Alí Alejo
- Subjects
0301 basic medicine ,viruses ,Immunology ,Orthomyxoviridae ,Genetic Vectors ,Pregnancy Proteins ,medicine.disease_cause ,Microbiology ,Virus ,law.invention ,Adenoviridae ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Orthomyxoviridae Infections ,Interferon ,law ,Virology ,Vaccines and Antiviral Agents ,medicine ,Animals ,biology ,Genetic Therapy ,biology.organism_classification ,Survival Analysis ,Recombinant Proteins ,Interferon tau ,Disease Models, Animal ,030104 developmental biology ,Treatment Outcome ,Viral replication ,030220 oncology & carcinogenesis ,Insect Science ,Interferon Type I ,Recombinant DNA ,Interferon type I ,medicine.drug - Abstract
Ovine interferon tau (IFN-τ) is a unique type I interferon with low toxicity and a broad host range in vivo . We report the generation of a nonreplicative recombinant adenovirus expressing biologically active IFN-τ. Using the B6.A2G-Mx1 mouse model, we showed that single-dose intranasal administration of recombinant Ad5-IFN-τ can effectively prevent lethality and disease induced by highly virulent hv-PR8 influenza virus by activating the interferon response and preventing viral replication.
- Published
- 2016
45. Use of alternative receptors different than α-dystroglycan by selected isolates of lymphocytic choriomeningitis virus
- Author
-
Michael B. A. Oldstone, Noemí Sevilla, Stefan Kunz, and Jillian M. Rojek
- Subjects
α dystroglycan ,Plasma protein binding ,In Vitro Techniques ,Lymphocytic choriomeningitis ,Virus ,Dystroglycans ,Mice ,Viral Proteins ,03 medical and health sciences ,Immune system ,Virology ,Dystroglycan ,medicine ,Animals ,Lymphocytic choriomeningitis virus ,Receptor ,Cells, Cultured ,Glycoproteins ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Membrane Glycoproteins ,biology ,030302 biochemistry & molecular biology ,medicine.disease ,3. Good health ,Cytoskeletal Proteins ,biology.protein ,Receptors, Virus ,Protein Processing, Post-Translational ,Protein Binding - Abstract
Long-term infections with viruses permit the generation of variants that evolve specific growth advantages in certain tissues and may show altered disease potentials. The selection of such variants is influenced by the host tissue and often involves virus-receptor interactions. Here we report studies of receptor usage by several lymphocytic choriomeningitis virus (LCMV) isolates that expressed different disease patterns. Consistent with our previous studies, we found that, with one exception, multiple LCMV variants that cause suppression of immune responses bound with high affinity to their cellular receptor alpha-dystroglycan (alpha-DG) and were dependent on alpha-DG for entry and infection. The exception also bound strongly to alpha-DG but was not dependent on alpha-DG for entry and infection. In contrast, those variants of LCMV that do not suppress the immune response either displayed low or no binding affinity for alpha-DG and used alternative receptors in addition to or instead of alpha-DG for entry and infection. For all alpha-DG binding variants, alpha-DG represents the preferred receptor in DG-expressing cells, as soluble alpha-DG blocked their infection of DG-deficient cells, indicating that binding of alpha-DG to the viral glycoprotein (GP) at the virion surface interferes with the GP's interaction with the alternative receptor. Biochemical characterization of the alternative receptor(s) for LCMV indicated that they are either protein(s) or protein-bound entities.
- Published
- 2004
- Full Text
- View/download PDF
46. Viral targeting of hematopoietic progenitors and inhibition of DC maturation as a dual strategy for immune subversion
- Author
-
Chao Teng, Dorian B. McGavern, Michael B. A. Oldstone, Noemí Sevilla, and Stefan Kunz
- Subjects
Time Factors ,viruses ,CD8 Antigens ,T-Lymphocytes ,Bone Marrow Cells ,chemical and pharmacologic phenomena ,Lymphocytic Choriomeningitis ,Lymphocytic choriomeningitis ,Models, Biological ,Article ,Mice ,Immune system ,MHC class I ,Immune Tolerance ,medicine ,Humans ,Animals ,Lymphocytic choriomeningitis virus ,Progenitor cell ,Immunosuppression Therapy ,CD86 ,MHC class II ,Microscopy, Confocal ,biology ,Membrane Proteins ,hemic and immune systems ,Dendritic Cells ,General Medicine ,Flow Cytometry ,Hematopoietic Stem Cells ,medicine.disease ,Immunohistochemistry ,CD11c Antigen ,Cell biology ,Mice, Inbred C57BL ,Virus Diseases ,Immunology ,Commentary ,biology.protein ,Interferons ,Clone (B-cell biology) ,Immunosuppressive Agents ,Cell Division ,Spleen ,CD80 - Abstract
DCs play a pivotal role in bringing forth innate and adaptive immune responses. Viruses can specifically target DCs, rendering them ineffective in stimulating T cells, which can ultimately lead to immunosuppression. In the present study we have identified several potential mechanisms by which lymphocytic choriomeningitis virus (LCMV) induces immunosuppression in its natural murine host. The immunosuppressive LCMV variant clone 13 (Cl 13) infects DCs and interferes with their maturation and antigen-presenting capacity as evidenced by a significant reduction in the surface expression of MHC class I, MHC class II, CD40, CD80, and CD86 molecules. Additionally, Cl 13 infects hematopoietic progenitor cells both in vivo and in vitro, impairing their development. One mechanism by which hematopoietic progenitors are developmentally impaired is through the Cl 13-induced production of IFN-alpha and IFN-beta (IFN-alpha/beta). Mice deficient in the receptor for IFN-alpha/beta show a normal differentiation of progenitors into DCs despite viral infection. Thus, a virus can evolve a strategy to boost its survival by preventing the maturation of DCs from infected progenitor cells and by reducing the expression of antigen-presenting and costimulatory molecules on developed DCs.
- Published
- 2004
- Full Text
- View/download PDF
47. Protective Efficacy in Sheep of Adenovirus-Vectored Vaccines against Bluetongue Virus Is Associated with Specific T Cell Responses
- Author
-
Miguel Avia, Noemí Sevilla, Verónica Martín, Lourdes Peña, Elena Pascual, and F. Valcárcel
- Subjects
T-Lymphocytes ,T cell ,viruses ,lcsh:Medicine ,Viral Nonstructural Proteins ,medicine.disease_cause ,Bluetongue ,Virus ,Adenoviridae ,Mice ,Immune system ,Immunity ,Cricetinae ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,lcsh:Science ,Vero Cells ,Vaccines, Synthetic ,Sheep ,Multidisciplinary ,Orbivirus ,biology ,Viral Vaccine ,lcsh:R ,Viral Vaccines ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Virology ,Mice, Inbred C57BL ,Vaccination ,HEK293 Cells ,medicine.anatomical_structure ,Immunology ,Capsid Proteins ,Female ,lcsh:Q ,Research Article - Abstract
Bluetongue virus (BTV) is an economically important Orbivirus of the Reoviridae family that causes a hemorrhagic disease in ruminants. Its control has been achieved by inactivatedvaccines that have proven to protect against homologous BTV challenge although unable to induce long-term immunity. Therefore, a more efficient control strategy needs to be developed. Recombinant adenovirus vectors are lead vaccine candidates for protection of several diseases, mainly because of their potency to induce potent T cell immunity. Here we report the induction of humoral and T-cell mediated responses able to protect animals against BTV challenge by recombinant replication-defective human adenovirus serotype 5 (Ad5) expressing either VP7, VP2 or NS3 BTV proteins. First we used the IFNAR(-/-) mouse model system to establish a proof of principle, and afterwards we assayed the protective efficacy in sheep, the natural host of BTV. Mice were completely protected against BTV challenge, developing humoral and BTV-specific CD8+- and CD4+-T cell responses by vaccination with the different rAd5. Sheep vaccinated with Ad5-BTV-VP2 and Ad5-BTV-VP7 or only with Ad5-BTV-VP7 and challenged with BTV showed mild disease symptoms and reduced viremia. This partial protection was achieved in the absence of neutralizing antibodies but strong BTV-specific CD8+ T cell responses in those sheep vaccinated with Ad5- BTV-VP7. These data indicate that rAd5 is a suitable vaccine vector to induce T cell immunity during BTV vaccination and provide new data regarding the relevance of T cell responses in protection during BTV infection. © 2015 Martín et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
- Published
- 2015
48. Ovine and murine T cell epitopes from the non-structural protein 1 (NS1) of bluetongue virus serotype 8 (BTV-8) are shared among viral serotypes
- Author
-
Noemí Sevilla, Verónica Martín, Lourdes Peña, José M. Rojas, Ministerio de Economía y Competitividad (España), and European Commission
- Subjects
CD4-Positive T-Lymphocytes ,T cell ,[SDV]Life Sciences [q-bio] ,Epitopes, T-Lymphocyte ,Peptide binding ,chemical and pharmacologic phenomena ,CD8-Positive T-Lymphocytes ,Cross Reactions ,Viral Nonstructural Proteins ,Bluetongue ,Epitope ,Virus ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Cytotoxic T cell ,030304 developmental biology ,0303 health sciences ,Sheep ,Orbivirus ,General Veterinary ,biology ,Research ,ELISPOT ,biology.organism_classification ,Virology ,veterinary(all) ,3. Good health ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Female ,Bluetongue virus ,030215 immunology - Abstract
Bluetongue virus (BTV) is a non-enveloped dsRNA virus that causes a haemorrhagic disease mainly in sheep. It is an economically important Orbivirus of the Reoviridae family. In order to estimate the importance of T cell responses during BTV infection, it is essential to identify the epitopes targeted by the immune system. In the present work, we selected potential T cell epitopes (3 MHC-class II-binding and 8 MHC-class I binding peptides) for the C57BL/6 mouse strain from the BTV-8 non-structural protein NS1, using H2b-binding predictive algorithms. Peptide binding assays confirmed all MHC-class I predicted peptides bound MHC-class I molecules. The immunogenicity of these 11 predicted peptides was then determined using splenocytes from BTV-8-inoculated C57BL/6 mice. Four MHC-class I binding peptides elicited specific IFN-γ production and generated cytotoxic T lymphocytes (CTL) in BTV-8 infected mice. CTL specific for 2 of these peptides were also able to recognise target cells infected with different BTV serotypes. Similarly, using a combination of IFN-γ ELISPOT, intracellular cytokine staining and proliferation assays, two MHC-class II peptides were identified as CD4+ T cell epitopes in BTV-8 infected mice. Importantly, two peptides were also consistently immunogenic in sheep infected with BTV-8 using IFN-γ ELISPOT assays. Both of these peptides stimulated CD4+ T cells that cross-reacted with other BTV serotypes. The characterisation of these T cell epitopes can help develop vaccines protecting against a broad spectrum of BTV serotypes and differentiate infected from vaccinated animals., This work was funded by grants AGL2009-07353, RyC-2010-06516 and AGL2011-25025 from Ministerio de Economía y Competitividad (Spain) and 228394-NADIR Integrating Activities 7th EU program.
- Published
- 2014
- Full Text
- View/download PDF
49. Molecular analysis of the interaction of LCMV with its cellular receptor α-dystroglycan
- Author
-
Dorian B. McGavern, Kevin P. Campbell, Noemí Sevilla, Michael B. A. Oldstone, and Stefan Kunz
- Subjects
Protein subunit ,viruses ,Biology ,Lymphocytic choriomeningitis ,Binding, Competitive ,Article ,03 medical and health sciences ,Mice ,Protein structure ,Laminin ,medicine ,Animals ,Arenaviridae Infections ,Lymphocytic choriomeningitis virus ,Binding site ,Dystroglycans ,Actin ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,Extracellular Matrix Proteins ,Binding Sites ,Membrane Glycoproteins ,030302 biochemistry & molecular biology ,Cell Biology ,medicine.disease ,Molecular biology ,Transmembrane protein ,3. Good health ,Cell biology ,Protein Structure, Tertiary ,Mice, Inbred C57BL ,Cytoskeletal Proteins ,Tissue tropism ,biology.protein ,lymphocytic choriomeningitis virus ,viral receptor ,binding site ,extracellular matrix ,pathogenesis ,Female ,Spleen - Abstract
alpha-Dystroglycan (DG) has been identified as the cellular receptor for lymphocytic choriomeningitis virus (LCMV) and Lassa fever virus (LFV). This subunit of DG is a highly versatile cell surface molecule that provides a molecular link between the extracellular matrix (ECM) and a beta-DG transmembrane component, which interacts with the actin-based cytoskeleton. In addition, DG exhibits a complex pattern of interaction with a wide variety of ECM and cellular proteins. In the present study, we characterized the binding of LCMV to alpha-DG and addressed the role of alpha-DG-associated host-derived proteins in virus infection. We found that the COOH-terminal region of alpha-DG's first globular domain and the NH2-terminal region of the mucin-related structures of alpha-DG together form the binding site for LCMV. The virus-alpha-DG binding unlike ECM alpha-DG interactions was not dependent on divalent cations. Despite such differences in binding, LCMV and laminin-1 use, in part, an overlapping binding site on alpha-DG, and the ability of an LCMV isolate to compete with laminin-1 for receptor binding is determined by its binding affinity to alpha-DG. This competition of the virus with ECM molecules for receptor binding likely explains the recently found correlation between the affinity of LCMV binding to alpha-DG, tissue tropism, and pathological potential. LCMV strains and variants with high binding affinity to alpha-DG but not low affinity binders are able to infect CD11c+ dendritic cells, which express alpha-DG at their surface. Infection followed by dysfunction of these antigen-presenting cells contributes to immunosuppression and persistent viral infection in vivo.
- Published
- 2001
50. Immunosuppression and Resultant Viral Persistence by Specific Viral Targeting of Dendritic Cells
- Author
-
Noemí Sevilla, Juan Carlos de la Torre, Andreas Holz, Stefan Kunz, Hiroki Yamada, Kevin P. Campbell, Michael B. A. Oldstone, Dirk Homann, and Hanna Lewicki
- Subjects
Central Nervous System ,CD11c+/DEC-205+ cells ,viruses ,Virus Replication ,Mice ,0302 clinical medicine ,Cricetinae ,Lymphocytic choriomeningitis virus ,Immunology and Allergy ,Cytotoxic T cell ,Dystroglycans ,In Situ Hybridization ,Mice, Knockout ,Mice, Inbred BALB C ,0303 health sciences ,Membrane Glycoproteins ,tropism ,3. Good health ,Receptors, Virus ,Original Article ,Tumor necrosis factor alpha ,viral receptor ,Protein Binding ,Immunology ,selection ,Receptors, Cell Surface ,Lymphocytic Choriomeningitis ,Biology ,Lymphocytic choriomeningitis ,Virus ,Cell Line ,Minor Histocompatibility Antigens ,Viral Proteins ,03 medical and health sciences ,Immune system ,Antigens, CD ,Viral entry ,medicine ,Animals ,Lectins, C-Type ,030304 developmental biology ,Immunosuppression Therapy ,CD11 Antigens ,Tumor Necrosis Factor-alpha ,Dendritic Cells ,medicine.disease ,Virology ,Cytoskeletal Proteins ,Viral replication ,Cell culture ,Chronic Disease ,Spleen ,T-Lymphocytes, Cytotoxic ,030215 immunology - Abstract
Among cells of the immune system, CD11c(+) and DEC-205(+) splenic dendritic cells primarily express the cellular receptor (alpha-dystroglycan [alpha-DG]) for lymphocytic choriomeningitis virus (LCMV). By selection, strains and variants of LCMV that bind alpha-DG with high affinity are associated with virus replication in the white pulp, show preferential replication in a majority of CD11c(+) and DEC-205(+) cells, cause immunosuppression, and establish a persistent infection. In contrast, viral strains and variants that bind with low affinity to alpha-DG are associated with viral replication in the red pulp, display minimal replication in CD11c(+) and DEC-205(+) cells, and generate a robust anti-LCMV cytotoxic T lymphocyte response that clears the virus infection. Differences in binding affinities can be mapped to a single amino acid change in the viral glycoprotein 1 ligand that binds to alpha-DG. These findings indicate that receptor-virus interaction on dendritic cells in vivo can be an essential step in the initiation of virus-induced immunosuppression and viral persistence.
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.