Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 44(2): 233-45, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26872694

RESUMEN

According to in vitro assays, T cells are thought to kill rapidly and efficiently, but the efficacy and dynamics of cytotoxic T lymphocyte (CTL)-mediated killing of virus-infected cells in vivo remains elusive. We used two-photon microscopy to quantify CTL-mediated killing in mice infected with herpesviruses or poxviruses. On average, one CTL killed 2-16 virus-infected cells per day as determined by real-time imaging and by mathematical modeling. In contrast, upon virus-induced MHC class I downmodulation, CTLs failed to destroy their targets. During killing, CTLs remained migratory and formed motile kinapses rather than static synapses with targets. Viruses encoding the calcium sensor GCaMP6s revealed strong heterogeneity in individual CTL functional capacity. Furthermore, the probability of death of infected cells increased for those contacted by more than two CTLs, indicative of CTL cooperation. Thus, direct visualization of CTLs during killing of virus-infected cells reveals crucial parameters of CD8(+) T cell immunity.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Muromegalovirus/inmunología , Perforina/metabolismo , Subgrupos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Virus Vaccinia/inmunología , Vaccinia/inmunología , Animales , Señalización del Calcio , Comunicación Celular , Células Cultivadas , Citotoxicidad Inmunológica , Humanos , Evasión Inmune , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía de Fluorescencia por Excitación Multifotónica , Perforina/genética , Subgrupos de Linfocitos T/virología , Linfocitos T Citotóxicos/virología
2.
Mol Ther ; 32(7): 2406-2422, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38734899

RESUMEN

Vaccinia viruses (VACVs) are versatile therapeutic agents and different features of various VACV strains allow for a broad range of therapeutic applications. Modified VACV Ankara (MVA) is a particularly altered VACV strain that is highly immunogenic, incapable of replicating in mammalian hosts, and broadly used as a safe vector for vaccination. Alternatively, Western Reserve (WR) or Copenhagen (Cop) are VACV strains that efficiently replicate in cancer cells and, therefore, are used to develop oncolytic viruses. However, the immune evasion capacity of WR or Cop hinders their ability to elicit antitumor immune responses, which is crucial for efficacy in the clinic. Here, we describe a new VACV strain named Immune-Oncolytic VACV Ankara (IOVA), which combines efficient replication in cancer cells with induction of immunogenic tumor cell death (ICD). IOVA was engineered from an MVA ancestor and shows superior cytotoxicity in tumor cells. In addition, the IOVA genome incorporates mutations that lead to massive fusogenesis of tumor cells, which contributes to improved antitumor effects. In syngeneic mouse tumor models, the induction of ICD results in robust antitumor immunity directed against tumor neo-epitopes and eradication of large established tumors. These data present IOVA as an improved immunotherapeutic oncolytic vector.


Asunto(s)
Muerte Celular Inmunogénica , Viroterapia Oncolítica , Virus Oncolíticos , Virus Vaccinia , Virus Vaccinia/genética , Virus Vaccinia/inmunología , Animales , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Ratones , Humanos , Viroterapia Oncolítica/métodos , Línea Celular Tumoral , Neoplasias/terapia , Neoplasias/inmunología , Replicación Viral , Vectores Genéticos/genética
3.
J Infect Dis ; 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38195212

RESUMEN

Licensed vaccines against the Middle East respiratory syndrome coronavirus (MERS-CoV), an emerging pathogen of concern, are lacking. The Modified Vaccinia virus Ankara vector-based vaccine MVA-MERS-S, expressing the MERS-CoV-spike glycoprotein (MERS-S), is one of three candidate vaccines in clinical development and elicits robust humoral and cellular immunity. Here, we identified for the first time a MERS-S-specific CD8+ T-cell epitope in an HLA-A*03:01/HLA-B*35:01-positive vaccinee using a screening assay, intracellular cytokine staining, and in silico epitope prediction. As evidence from MERS-CoV infection suggests a protective role of long-lasting CD8+ T-cell responses, the identification of epitopes will facilitate longitudinal analyses of vaccine-induced T-cell immunity.

4.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34162739

RESUMEN

Severe acute respiratory syndrome (SARS) coronavirus 2 (SARS-CoV-2) has emerged as the infectious agent causing the pandemic coronavirus disease 2019 (COVID-19) with dramatic consequences for global human health and economics. Previously, we reached clinical evaluation with our vector vaccine based on modified vaccinia virus Ankara (MVA) against the Middle East respiratory syndrome coronavirus (MERS-CoV), which causes an infection in humans similar to SARS and COVID-19. Here, we describe the construction and preclinical characterization of a recombinant MVA expressing full-length SARS-CoV-2 spike (S) protein (MVA-SARS-2-S). Genetic stability and growth characteristics of MVA-SARS-2-S, plus its robust expression of S protein as antigen, make it a suitable candidate vaccine for industrial-scale production. Vaccinated mice produced S-specific CD8+ T cells and serum antibodies binding to S protein that neutralized SARS-CoV-2. Prime-boost vaccination with MVA-SARS-2-S protected mice sensitized with a human ACE2-expressing adenovirus from SARS-CoV-2 infection. MVA-SARS-2-S is currently being investigated in a phase I clinical trial as aspirant for developing a safe and efficacious vaccine against COVID-19.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Vacunas contra la COVID-19/inmunología , COVID-19/prevención & control , Glicoproteína de la Espiga del Coronavirus/inmunología , Animales , Vacunas contra la COVID-19/normas , Relación Dosis-Respuesta Inmunológica , Humanos , Ratones , Ratones Endogámicos BALB C , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/genética , Linfocitos T , Vacunación , Virus Vaccinia
5.
J Infect Dis ; 228(5): 586-590, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-36857443

RESUMEN

Modified vaccinia virus Ankara (MVA) is used as a vaccine against monkeypox virus and as a viral vaccine vector. MVA-MERS-S is a vaccine candidate against Middle East respiratory syndrome (MERS)-associated coronavirus. Here, we report that cross-reactive monkeypox virus neutralizing antibodies were detectable in only a single study participant after the first dose of MVA-MERS-S vaccine, in 3 of 10 after the second dose, and in 10 of 10 after the third dose.


Asunto(s)
Infecciones por Coronavirus , Coronavirus del Síndrome Respiratorio de Oriente Medio , Vacunas Virales , Humanos , Anticuerpos ampliamente neutralizantes , Glicoproteína de la Espiga del Coronavirus , Monkeypox virus , Anticuerpos Antivirales , Virus Vaccinia/genética , Infecciones por Coronavirus/prevención & control , Anticuerpos Neutralizantes
6.
Emerg Infect Dis ; 29(6): 1236-1239, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37209676

RESUMEN

We developed an ELISPOT assay for evaluating Middle East respiratory syndrome coronavirus (MERS-CoV)‒specific T-cell responses in dromedary camels. After single modified vaccinia virus Ankara-MERS-S vaccination, seropositive camels showed increased levels of MERS-CoV‒specific T cells and antibodies, indicating suitability of camel vaccinations in disease-endemic areas as a promising approach to control infection.


Asunto(s)
Camelus , Infecciones por Coronavirus , Linfocitos T , Vacunas Virales , Animales , Camelus/inmunología , Linfocitos T/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/veterinaria , Vacunas Virales/inmunología , Vacunación/veterinaria , Ensayo de Immunospot Ligado a Enzimas , Anticuerpos Antivirales
7.
PLoS Pathog ; 16(2): e1008279, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32023327

RESUMEN

IFN-γ is an enigmatic cytokine that shows direct anti-viral effects, confers upregulation of MHC-II and other components relevant for antigen presentation, and that adjusts the composition and balance of complex cytokine responses. It is produced during immune responses by innate as well as adaptive immune cells and can critically affect the course and outcome of infectious diseases, autoimmunity, and cancer. To selectively analyze the function of innate immune cell-derived IFN-γ, we generated conditional IFN-γOFF mice, in which endogenous IFN-γ expression is disrupted by a loxP flanked gene trap cassette inserted into the first intron of the IFN-γ gene. IFN-γOFF mice were intercrossed with Ncr1-Cre or CD4-Cre mice that express Cre mainly in NK cells (IFN-γNcr1-ON mice) or T cells (IFN-γCD4-ON mice), respectively. Rosa26RFP reporter mice intercrossed with Ncr1-Cre mice showed selective RFP expression in more than 80% of the NK cells, while upon intercrossing with CD4-Cre mice abundant RFP expression was detected in T cells, but also to a minor extent in other immune cell subsets. Previous studies showed that IFN-γ expression is needed to promote survival of vaccinia virus (VACV) infection. Interestingly, during VACV infection of wild type and IFN-γCD4-ON mice two waves of serum IFN-γ were induced that peaked on day 1 and day 3/4 after infection. Similarly, VACV infected IFN-γNcr1-ON mice mounted two waves of IFN-γ responses, of which the first one was moderately and the second one profoundly reduced when compared with WT mice. Furthermore, IFN-γNcr1-ON as well as IFN-γCD4-ON mice survived VACV infection, whereas IFN-γOFF mice did not. As expected, ex vivo analysis of splenocytes derived from VACV infected IFN-γNcr1-ON mice showed IFN-γ expression in NK cells, but not T cells, whereas IFN-γOFF mice showed IFN-γ expression neither in NK cells nor T cells. VACV infected IFN-γNcr1-ON mice mounted normal cytokine responses, restored neutrophil accumulation, and showed normal myeloid cell distribution in blood and spleen. Additionally, in these mice normal MHC-II expression was detected on peripheral macrophages, whereas IFN-γOFF mice did not show MHC-II expression on such cells. In conclusion, upon VACV infection Ncr1 positive cells including NK cells mount two waves of early IFN-γ responses that are sufficient to promote the induction of protective anti-viral immunity.


Asunto(s)
Antígenos Ly/inmunología , Regulación de la Expresión Génica/inmunología , Interferón gamma/inmunología , Células Asesinas Naturales/inmunología , Receptor 1 Gatillante de la Citotoxidad Natural/inmunología , Virus Vaccinia/inmunología , Vaccinia/inmunología , Animales , Antígenos Ly/genética , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Interferón gamma/genética , Células Asesinas Naturales/patología , Ratones , Ratones Transgénicos , Receptor 1 Gatillante de la Citotoxidad Natural/genética , Linfocitos T/inmunología , Linfocitos T/patología , Vaccinia/genética , Vaccinia/patología , Virus Vaccinia/genética
8.
Emerg Infect Dis ; 27(7): 1974-1976, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34152973

RESUMEN

We report a therapy cat in a nursing home in Germany infected with severe acute respiratory syndrome coronavirus 2 during a cluster outbreak in the home residents. Although we confirmed prolonged presence of virus RNA in the asymptomatic cat, genome sequencing showed no further role of the cat in human infections on site.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Gatos , Brotes de Enfermedades , Alemania , Humanos , ARN Viral/genética , Jubilación
9.
Ann Rheum Dis ; 80(12): 1537-1544, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34226189

RESUMEN

OBJECTIVES: The monoclonal anti-CD20 antibody rituximab is frequently applied in the treatment of lymphoma as well as autoimmune diseases and confers efficient depletion of recirculating B cells. Correspondingly, B cell-depleted patients barely mount de novo antibody responses during infections or vaccinations. Therefore, efficient immune responses of B cell-depleted patients largely depend on protective T cell responses. METHODS: CD8+ T cell expansion was studied in rituximab-treated rheumatoid arthritis (RA) patients and B cell-deficient mice on vaccination/infection with different vaccines/pathogens. RESULTS: Rituximab-treated RA patients vaccinated with Influvac showed reduced expansion of influenza-specific CD8+ T cells when compared with healthy controls. Moreover, B cell-deficient JHT mice infected with mouse-adapted Influenza or modified vaccinia virus Ankara showed less vigorous expansion of virus-specific CD8+ T cells than wild type mice. Of note, JHT mice do not have an intrinsic impairment of CD8+ T cell expansion, since infection with vaccinia virus induced similar T cell expansion in JHT and wild type mice. Direct type I interferon receptor signalling of B cells was necessary to induce several chemokines in B cells and to support T cell help by enhancing the expression of MHC-I. CONCLUSIONS: Depending on the stimulus, B cells can modulate CD8+ T cell responses. Thus, B cell depletion causes a deficiency of de novo antibody responses and affects the efficacy of cellular response including cytotoxic T cells. The choice of the appropriate vaccine to vaccinate B cell-depleted patients has to be re-evaluated in order to efficiently induce protective CD8+ T cell responses.


Asunto(s)
Antirreumáticos/efectos adversos , Artritis Reumatoide/tratamiento farmacológico , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunogenicidad Vacunal/inmunología , Vacunas contra la Influenza/inmunología , Interferón Tipo I/inmunología , Rituximab/efectos adversos , Animales , Estudios de Casos y Controles , Citocinas/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Vacunas contra la Influenza/uso terapéutico , Gripe Humana/prevención & control , Ratones , Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/inmunología , Vaccinia/inmunología , Virus Vaccinia/inmunología
10.
Virol J ; 18(1): 70, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33827614

RESUMEN

BACKGROUND: The porcine circovirus type 2 (PCV2) is divided into eight genotypes including the previously described genotypes PCV2a to PCV2f and the two new genotypes PCV2g and PCV2h. PCV2 genotyping has become an important task in molecular epidemiology and to advance research on the prophylaxis and pathogenesis of PCV2 associated diseases. Standard genotyping of PCV2 is based on the sequencing of the viral genome or at least of the open reading frame 2. Although, the circovirus genome is small, classical sequencing is time consuming, expensive, less sensitive and less compatible with mass testing compared with modern real-time PCR assays. Here we report about a new PCV2 genotyping method using qPCR. METHODS: Based on the analysis of several hundred PCV2 full genome sequences, we identified PCV2 genotype specific sequences or single-nucleotide polymorphisms. We designed six TaqMan PCR assays that are specific for single genotypes PCV2a to PCV2f and two qPCRs targeting two genotypes simultaneously (PCV2g/PCV2d and PCV2h/PCV2c). To improve specific binding of oligonucleotide primers and TaqMan probes, we used locked nucleic acid technology. We evaluated amplification efficiency, diagnostic sensitivity and tested assay specificity for the respective genotypes. RESULTS: All eight PCV2 genotype specific qPCRs demonstrated appropriate amplification efficiencies between 91 and 97%. Testing samples from an epidemiological field study demonstrated a diagnostic sensitivity of the respective genotype specific qPCR that was comparable to a highly sensitive pan-PCV2 qPCR system. Genotype specificity of most qPCRs was excellent. Limited unspecific signals were obtained when a high viral load of PCV2b was tested with qPCRs targeting PCV2d or PCV2g. The same was true for the PCV2a specific qPCR when high copy numbers of PCV2d were tested. The qPCR targeting PCV2h/PCV2c showed some minor cross-reaction with PCV2d, PCV2f and PCV2g. CONCLUSION: Genotyping of PCV2 is important for routine diagnosis as well as for epidemiological studies. The introduced genotyping qPCR system is ideal for mass testing and should be a valuable complement to PCV2 sequencing, especially in the case of simultaneous infections with multiple PCV2 genotypes, subclinically infected animals or research studies that require large sample numbers.


Asunto(s)
Infecciones por Circoviridae , Circovirus , Enfermedades de los Porcinos , Animales , Infecciones por Circoviridae/diagnóstico , Infecciones por Circoviridae/veterinaria , Circovirus/genética , Genotipo , Filogenia , Reacción en Cadena en Tiempo Real de la Polimerasa , Porcinos , Enfermedades de los Porcinos/diagnóstico , Enfermedades de los Porcinos/virología
11.
Eur J Immunol ; 48(12): 2042-2054, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30259962

RESUMEN

"Immunogenic cell death" (ICD) is associated with the emission of so-called damage-associated molecular patterns (DAMPs) which trigger the immune response against dead-cell associated antigens. The secretion of the DAMP, adenosine triphosphate (ATP) has been shown to be autophagy-dependent. Here, we demonstrate that Modified Vaccinia virus Ankara (MVA), a highly attenuated strain of vaccinia virus, induces both cell death and autophagy in murine bone marrow-derived dendritic cells (BMDCs), which in turn confer the (cross-)priming of OVA-specific cytotoxic T cells (OT-I cells). Additionally, we show that MVA infection leads to increased extracellular ATP (eATP) as well as intracellular ATP (iATP) levels, with the latter being influenced by the autophagy. Furthermore, we show that the increased eATP supports the proliferation of OT-I cells and inhibition of the P2RX7 receptors results in an abrogation of the proliferation. These data reveal novel mechanisms on how MVA enhances adaptive immunity in vaccine strategies.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Viruela/inmunología , Virus Vaccinia/inmunología , Vacunas Virales/inmunología , Inmunidad Adaptativa , Adenosina Trifosfato/inmunología , Adenosina Trifosfato/metabolismo , Animales , Autofagia , Células de la Médula Ósea/inmunología , Muerte Celular , Proliferación Celular , Células Cultivadas , Reactividad Cruzada , Citotoxicidad Inmunológica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Purinérgicos P2X7/metabolismo , Vacunas de ADN
12.
J Infect Dis ; 218(4): 614-623, 2018 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-29912453

RESUMEN

Background: High-pathogenicity avian influenza viruses continue to circulate in poultry and wild birds and occasionally infect humans, sometimes with fatal outcomes. Development of vaccines is a priority to prepare for potential pandemics but is complicated by antigenic variation of the surface glycoprotein hemagglutinin. We report the immunological profile induced by human immunization with modified vaccinia virus Ankara (MVA) expressing the hemagglutinin gene of influenza A(H5N1) virus A/Vietnam/1194/04 (rMVA-H5). Methods: In a double-blinded phase 1/2a clinical trial, 79 individuals received 1 or 2 injections of rMVA-H5 or vector control. Twenty-seven study subjects received a booster immunization after 1 year. The breadth, magnitude, and properties of vaccine-induced antibody and T-cell responses were characterized. Results: rMVA-H5 induced broadly reactive antibody responses, demonstrated by protein microarray, hemagglutination inhibition, virus neutralization, and antibody-dependent cellular cytotoxicity assays. Antibodies cross-reacted with antigenically distinct H5 viruses, including the recently emerged subtypes H5N6 and H5N8 and the currently circulating subtype H5N1. In addition, the induction of T cells specific for H5 viruses of 2 different clades was demonstrated. Conclusions: rMVA-H5 induced immune responses that cross-reacted with H5 viruses of various clades. These findings validate rMVA-H5 as vaccine candidate against antigenically distinct H5 viruses. Clinical Trials Registration: NTR3401.


Asunto(s)
Anticuerpos Antivirales/sangre , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Linfocitos T/inmunología , Adulto , Citotoxicidad Celular Dependiente de Anticuerpos , Reacciones Cruzadas , Método Doble Ciego , Portadores de Fármacos , Femenino , Pruebas de Inhibición de Hemaglutinación , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Humanos , Esquemas de Inmunización , Subtipo H5N1 del Virus de la Influenza A/genética , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Masculino , Pruebas de Neutralización , Análisis por Matrices de Proteínas , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Virus Vaccinia/genética , Adulto Joven
13.
J Hepatol ; 68(4): 682-690, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29274730

RESUMEN

BACKGROUND & AIM: Virus-induced fulminant hepatitis is a major cause of acute liver failure. During acute viral hepatitis the impact of type I interferon (IFN-I) on myeloid cells, including liver-resident Kupffer cells (KC), is only partially understood. Herein, we dissected the impact of locally induced IFN-I responses on myeloid cell function and hepatocytes during acute liver inflammation. METHODS: Two different DNA-encoded viruses, vaccinia virus (VACV) and murine cytomegalovirus (MCMV), were studied. In vivo imaging was applied to visualize local IFN-ß induction and IFN-I receptor (IFNAR) triggering in VACV-infected reporter mice. Furthermore, mice with a cell type-selective IFNAR ablation were analyzed to dissect the role of IFNAR signaling in myeloid cells and hepatocytes. Experiments with Cx3cr1+/gfp mice revealed the origin of reconstituted KC. Finally, mixed bone marrow chimeric mice were studied to specifically analyze the effect of IFNAR triggering on liver infiltrating monocytes. RESULTS: VACV infection induced local IFN-ß responses, which lead to IFNAR signaling primarily within the liver. IFNAR triggering was needed to control the infection and prevent fulminant hepatitis. The severity of liver inflammation was independent of IFNAR triggering of hepatocytes, whereas IFNAR triggering of myeloid cells protected from excessive inflammation. Upon VACV or MCMV infection KC disappeared, whereas infiltrating monocytes differentiated to KC afterwards. During IFNAR triggering such replenished monocyte-derived KC comprised more IFNAR-deficient than -competent cells in mixed bone marrow chimeric mice, whereas after the decline of IFNAR triggering both subsets showed an even distribution. CONCLUSION: Upon VACV infection IFNAR triggering of myeloid cells, but not of hepatocytes, critically modulates acute viral hepatitis. During infection with DNA-encoded viruses IFNAR triggering of liver-infiltrating blood monocytes delays the development of monocyte-derived KC, pointing towards new therapeutic strategies for acute viral hepatitis. LAY SUMMARY: Viral infection can cause fulminant hepatitis, which in turn is a major cause of acute liver failure. Herein, we aimed to study the role of type 1 interferon responses in acute viral hepatitis. We identified that during infection with DNA-encoded viruses, type 1 interferon receptor triggering of blood monocytes delays the development of monocyte-derived Kupffer cells. This points to new therapeutic strategies for acute viral hepatitis.


Asunto(s)
Hepatitis Viral Animal/fisiopatología , Macrófagos del Hígado/fisiología , Receptor de Interferón alfa y beta/fisiología , Transducción de Señal/fisiología , Enfermedad Aguda , Animales , Hepatitis Viral Animal/etiología , Ratones , Ratones Endogámicos C57BL , Vaccinia/fisiopatología
14.
Virol J ; 15(1): 25, 2018 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-29378597

RESUMEN

BACKGROUND: The occurrence of the novel porcine circovirus type 3 (PCV3) was reported from the Americas, Asia and Europe. Although this virus was detected in association with various clinical syndromes in pigs, its role as possible swine pathogen remains unclear. PCV3 was detected with high prevalence in Polish farms, but to date no genome sequences were available from European PCV3 strains. METHODS: We collected 1060 serum samples from piglets at the age of 20-24 weeks from 53 farms distributed all over Germany. PCV3 DNA was detected using a real-time PCR and subsequently complete PCV3 genome sequences were obtained after multiply primed rolling circle amplification and sequencing of overlapping PCR products. Phylogenetic analysis was performed by neighbor-joining method and maximum likelihood method. RESULTS: We obtained 15 complete PCV3 genome sequences as well as nine partial sequences including the putative ORFs 1, 2 and 3 from PCV3 viremic animals in German pig farms. Phylogenetic analysis of these German as well as 30 full genome sequences received from GenBank divided the PCV3 strains into two main groups and several subclusters. Furthermore, we were able to define group specific amino acid patterns in open reading frame 1 and 2. CONCLUSION: PCV3 is distributed with high prevalence in German pig industry. Phylogenetic analysis revealed two clearly separated groups of PCV3 strains, which might be considered as PCV3 genotypes. Specific nucleotide and amino acid marker positions may serve for easy and fast intraspecies classification and genotyping of PCV3 strains. No correlation between PCV3 variants with their geographical origin was evident. We found the same diversity of PCV3 strains in Germany as in other countries. We hypothesize that PCV3 is not a newly emerging virus in the German pig population. Future studies will have to show, if PCV3 genotype specific biological properties are evident.


Asunto(s)
Infecciones por Circoviridae/veterinaria , Circovirus/clasificación , Circovirus/genética , Genoma Viral , Genómica , Enfermedades de los Porcinos/virología , Animales , Genómica/métodos , Genotipo , Alemania/epidemiología , Sistemas de Lectura Abierta , Filogenia , Prevalencia , Análisis de Secuencia de ADN , Porcinos , Enfermedades de los Porcinos/epidemiología
15.
Biologicals ; 54: 58-62, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29759890

RESUMEN

Modified Vaccinia Virus Ankara (MVA) is a highly attenuated and replication-deficient virus serving as vaccine against infectious diseases. Here, we assessed the in vivo distribution of a recombinant MVA candidate vaccine against the Middle Eastern Respiratory Syndrome (MVA-MERS-S) in mice. Intramuscularly inoculated mice were necropsied at different time points and examined by histology, immunohistochemistry and real-time PCR. We detected inflammation and myonecrosis at the parenteral site and hyperplasia of the draining lymph nodes. MVA-MERS-S did not result in detectable lesions in tissues peripheral to the parenteral site and draining lymph nodes. Real-time PCR analysis of >240 tissue samples detected MVA-DNA predominantly at the injection site and in the draining lymph nodes, and suggested continuous clearance of the candidate vaccine during the observation period. Levels of parenteral site inflammation and hyperplasia of draining lymph nodes were considered in line with immunological responses to vaccine inoculation.


Asunto(s)
Infecciones por Coronavirus , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Vacunación , Virus Vaccinia/inmunología , Vacunas Virales , Animales , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/prevención & control , Inyecciones Intramusculares , Ratones , Vacunas Virales/inmunología , Vacunas Virales/farmacología
17.
J Gen Virol ; 98(8): 2113-2117, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28758619

RESUMEN

We identified a novel papillomavirus, Sus scrofa papillomavirus 2 (SsPV2), which is the first papillomavirus associated with papillomas in pigs. In skin alterations of a German wild boar, showing typical gross and histological appearance of papillomas, papillomavirus-like particles were demonstrated by electron microscopy. Degenerate papillomavirus-specific primers were used to amplify and sequence parts of the viral DNA. Subsequently, the complete genomic DNA was cloned and sequenced. The SsPV2 genome had a length of 8218 bp, encoded the early proteins E6, E7, E1 and E2, the late proteins L1 and L2 and contained an upstream regulatory region. Genomic characterization demonstrated papillomavirus-typical characteristics as well as unique features. For example, the E2 protein was significantly larger than in every other known papillomavirus species. Phylogenetic analysis was not able to relate SsPV2 unambiguously with other papillomavirus species or existing genera. Therefore, it might be representative of a new papillomavirus genus.


Asunto(s)
Papillomaviridae/aislamiento & purificación , Infecciones por Papillomavirus/veterinaria , Sus scrofa/virología , Enfermedades de los Porcinos/virología , Animales , Genoma Viral , Sistemas de Lectura Abierta , Papillomaviridae/clasificación , Papillomaviridae/genética , Infecciones por Papillomavirus/virología , Porcinos , Proteínas Virales/genética , Proteínas Virales/metabolismo
18.
J Virol ; 90(22): 10209-10219, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27581985

RESUMEN

Due to antigenic drift of influenza viruses, seasonal influenza vaccines need to be updated annually. These vaccines are based on predictions of strains likely to circulate in the next season. However, vaccine efficacy is greatly reduced in the case of a mismatch between circulating and vaccine strains. Furthermore, novel antigenically distinct influenza viruses are introduced into the human population from animal reservoirs occasionally and may cause pandemic outbreaks. To dampen the impact of seasonal and pandemic influenza, vaccines that induce broadly protective and long-lasting immunity are preferred. Because influenza virus-specific CD8+ T cells are directed mainly against relatively conserved internal proteins, like nucleoprotein (NP), they are highly cross-reactive and afford protection against infection with antigenically distinct influenza virus strains, so-called heterosubtypic immunity. Here, we used modified vaccinia virus Ankara (MVA) as a vaccine vector for the induction of influenza virus NP-specific CD8+ T cells. To optimize the induction of CD8+ T cell responses, we made several modifications to NP, aiming at retaining the protein in the cytosol or targeting it to the proteasome. We hypothesized that these strategies would increase antigen processing and presentation and thus improve the induction of CD8+ T cell responses. We showed that NP with increased degradation rates improved CD8+ T cell activation in vitro if the amount of antigen was limited or if CD8+ T cells were of low functional avidity. However, after immunization of C57BL/6 mice, no differences were detected between modified NP and wild-type NP (NPwt), since NPwt already induced optimal CD8+ T cell responses. IMPORTANCE: Due to the continuous antigenic drift of seasonal influenza viruses and the threat of a novel pandemic, there is a great need for the development of novel influenza vaccines that offer broadly protective immunity against multiple subtypes. CD8+ T cells can provide immunity against multiple subtypes of influenza viruses by the recognition of relatively conserved internal antigens. In this study, we aimed at optimizing the CD8+ T cell response to influenza A virus by making modifications to influenza A virus nucleoprotein (NP) expressed from the modified vaccinia virus Ankara (MVA) vaccine vector. These modifications resulted in increased antigen degradation, thereby producing elevated levels of peptides that can be presented on major histocompatibility complex (MHC) class I molecules to CD8+ T cells. Although we were unable to increase the NP-specific immune response in the mouse strain used, this approach may have benefits for vaccine development using less-immunogenic proteins.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Virus de la Influenza A/inmunología , Virus de la Influenza A/metabolismo , Activación de Linfocitos/inmunología , Proteínas de Unión al ARN/metabolismo , Proteínas del Núcleo Viral/metabolismo , Animales , Anticuerpos Antivirales/metabolismo , Antígenos Virales/inmunología , Línea Celular , Línea Celular Tumoral , Pollos , Reacciones Cruzadas/inmunología , Perros , Femenino , Células HeLa , Humanos , Vacunas contra la Influenza/inmunología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos C57BL , Proteínas de la Nucleocápside , Infecciones por Orthomyxoviridae/virología , Proteolisis , Proteínas de Unión al ARN/inmunología , Vacunación/métodos , Virus Vaccinia/inmunología , Proteínas del Núcleo Viral/inmunología
19.
J Immunol ; 194(3): 1164-8, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25548218

RESUMEN

Efficient leukocyte migration is important for an effective host response to viral infection and the development of adaptive immunity. The poxvirus strain modified vaccinia virus Ankara (MVA), a safe and efficient viral vector, rapidly induces chemokine expression and respiratory recruitment of leukocytes, which is unique among vaccinia viruses. In addition to chemokines, the complement system contributes to the attraction and activation of different types of leukocytes. Using a murine model of intranasal infection, we show in this study that MVA-induced neutrophil recruitment depends on complement component C5. Remarkably, we find that C5 mediates neutrophil recruitment to the lung, even in the absence of the central complement component C3. Our findings argue for complement C5 activation during MVA infection of the lung via a C3-independent pathway, which enables rapid recruitment of neutrophils.


Asunto(s)
Complemento C5/inmunología , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Infecciones del Sistema Respiratorio/inmunología , Virus Vaccinia/inmunología , Animales , Quimiotaxis de Leucocito/inmunología , Complemento C3/genética , Complemento C3/inmunología , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/virología
20.
J Gen Virol ; 97(1): 128-133, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26555294

RESUMEN

We describe a novel papillomavirus - Rusa alfredi papillomavirus 1 (RalPV1) - which causes endemic fibropapillomatosis in the European conservation breeding population of the highly endangered Visayan spotted deer (Rusa alfredi). Degenerated papillomavirus-specific primers were used to amplify and sequence parts of the viral DNA. Subsequently, the complete genomic DNA was cloned and the sequence was determined. The RalPV1 genome has a length of 8029 bp, encodes the early proteins E6, E7, E1, E2 and E5, the two late proteins L1 and L2 and contains an upstream regulatory region. Highest sequence identities were observed with two deltapapillomaviruses, the Capreolus capreolus PV1 and Cervus elaphus PV1. Pairwise comparisons and phylogenetic analysis based on the ORF L1 suggested that RalPV1 is a putative new type of the papillomavirus species Deltapapillomavirus 5.


Asunto(s)
Ciervos/virología , Deltapapillomavirus/clasificación , Deltapapillomavirus/aislamiento & purificación , Enfermedades Endémicas , Papiloma/veterinaria , Animales , Análisis por Conglomerados , ADN Viral/química , ADN Viral/genética , Europa (Continente)/epidemiología , Genoma Viral , Histocitoquímica , Microscopía , Datos de Secuencia Molecular , Papiloma/epidemiología , Papiloma/patología , Papiloma/virología , Filogenia , Análisis de Secuencia de ADN , Homología de Secuencia , Proteínas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA