Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Immunology ; 154(2): 285-297, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29281850

RESUMEN

The immunological outcome of infections and vaccinations is largely determined during the initial first days in which antigen-presenting cells instruct T cells to expand and differentiate into effector and memory cells. Besides the essential stimulation of the T-cell receptor complex a plethora of co-stimulatory signals not only ensures a proper T-cell activation but also instils phenotypic and functional characteristics in the T cells appropriate to fight off the invading pathogen. The tumour necrosis factor receptor/ligand pair CD27/CD70 gained a lot of attention because of its key role in regulating T-cell activation, survival, differentiation and maintenance, especially in the course of viral infections and cancer. We sought to investigate the role of CD70 co-stimulation for immune responses induced by the vaccine vector modified vaccinia virus Ankara-Bavarian Nordic® (MVA-BN® ). Short-term blockade of CD70 diminished systemic CD8 T-cell effector and memory responses in mice. The dependence on CD70 became even more apparent in the lungs of MHC class II-deficient mice. Importantly, genetically encoded CD70 in MVA-BN® not only increased CD8 T-cell responses in wild-type mice but also substituted for CD4 T-cell help. MHC class II-deficient mice that were immunized with recombinant MVA-CD70 were fully protected against a lethal virus infection, whereas MVA-BN® -immunized mice failed to control the virus. These data are in line with CD70 playing an important role for vaccine-induced CD8 T-cell responses and prove the potency of integrating co-stimulatory molecules into the MVA-BN® backbone.


Asunto(s)
Ligando CD27/inmunología , Linfocitos T CD8-positivos/inmunología , Vectores Genéticos , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Inmunidad , Virus Vaccinia , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Biomarcadores , Ligando CD27/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Inmunización , Ratones , Ratones Noqueados , Virus Vaccinia/genética , Virus Vaccinia/inmunología
2.
J Virol ; 91(11)2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28331098

RESUMEN

There are currently no approved therapeutics or vaccines to treat or protect against the severe hemorrhagic fever and death caused by Ebola virus (EBOV). Ebola virus-like particles (EBOV VLPs) consisting of the matrix protein VP40, the glycoprotein (GP), and the nucleoprotein (NP) are highly immunogenic and protective in nonhuman primates against Ebola virus disease (EVD). We have constructed a modified vaccinia virus Ankara-Bavarian Nordic (MVA-BN) recombinant coexpressing VP40 and GP of EBOV Mayinga and the NP of Taï Forest virus (TAFV) (MVA-BN-EBOV-VLP) to launch noninfectious EBOV VLPs as a second vaccine modality in the MVA-BN-EBOV-VLP-vaccinated organism. Human cells infected with either MVA-BN-EBOV-VLP or MVA-BN-EBOV-GP showed comparable GP expression levels and transport of complex N-glycosylated GP to the cell surface. Human cells infected with MVA-BN-EBOV-VLP produced large amounts of EBOV VLPs that were decorated with GP spikes but excluded the poxviral membrane protein B5, thus resembling authentic EBOV particles. The heterologous TAFV NP enhanced EBOV VP40-driven VLP formation with efficiency similar to that of the homologous EBOV NP in a transient-expression assay, and both NPs were incorporated into EBOV VLPs. EBOV GP-specific CD8 T cell responses were comparable between MVA-BN-EBOV-VLP- and MVA-BN-EBOV-GP-immunized mice. The levels of EBOV GP-specific neutralizing and binding antibodies, as well as GP-specific IgG1/IgG2a ratios induced by the two constructs, in mice were also similar, raising the question whether the quality rather than the quantity of the GP-specific antibody response might be altered by an EBOV VLP-generating MVA recombinant.IMPORTANCE The recent outbreak of Ebola virus (EBOV), claiming more than 11,000 lives, has underscored the need to advance the development of safe and effective filovirus vaccines. Virus-like particles (VLPs), as well as recombinant viral vectors, have proved to be promising vaccine candidates. Modified vaccinia virus Ankara-Bavarian Nordic (MVA-BN) is a safe and immunogenic vaccine vector with a large capacity to accommodate multiple foreign genes. In this study, we combined the advantages of VLPs and the MVA platform by generating a recombinant MVA-BN-EBOV-VLP that would produce noninfectious EBOV VLPs in the vaccinated individual. Our results show that human cells infected with MVA-BN-EBOV-VLP indeed formed and released EBOV VLPs, thus producing a highly authentic immunogen. MVA-BN-EBOV-VLP efficiently induced EBOV-specific humoral and cellular immune responses in vaccinated mice. These results are the basis for future advancements, e.g., by including antigens from various filoviral species to develop multivalent VLP-producing MVA-based filovirus vaccines.


Asunto(s)
Vacunas contra el Virus del Ébola/inmunología , Ebolavirus/aislamiento & purificación , Glicoproteínas/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Virus Vaccinia/genética , Virión/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Virus del Ébola/genética , Ebolavirus/genética , Ebolavirus/inmunología , Ebolavirus/fisiología , Glicoproteínas/genética , Humanos , Inmunoglobulina G/sangre , Ratones , Nucleoproteínas/genética , Nucleoproteínas/inmunología , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/inmunología , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/inmunología , Virión/fisiología
3.
Part Fibre Toxicol ; 14(1): 21, 2017 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-28645296

RESUMEN

BACKGROUND: Dendritic cells (DCs) are specialized first-line sensors of foreign materials invading the organism. These sentinel cells rely on pattern recognition receptors such as Nod-like or Toll-like receptors (TLRs) to launch immune reactions against pathogens, but also to mediate tolerance to self-antigens and, in the intestinal milieu, to nutrients and commensals. Since inappropriate DC activation contributes to inflammatory diseases and immunopathologies, a key question in the evaluation of orally ingested nanomaterials is whether their contact with DCs in the intestinal mucosa disrupts this delicate homeostatic balance between pathogen defense and tolerance. Here, we generated steady-state DCs by incubating hematopoietic progenitors with feline McDonough sarcoma-like tyrosine kinase 3 ligand (Flt3L) and used the resulting immature DCs to test potential biological responses against food-grade synthetic amorphous silica (SAS) representing a common nanomaterial generally thought to be safe. RESULTS: Interaction of immature and unprimed DCs with food-grade SAS particles and their internalization by endocytic uptake fails to elicit cytotoxicity and the release of interleukin (IL)-1α or tumor necrosis factor-α, which were identified as master regulators of acute inflammation in lung-related studies. However, the display of maturation markers on the cell surface shows that SAS particles activate completely immature DCs. Also, the endocytic uptake of SAS particles into these steady-state DCs leads to induction of the pro-IL-1ß precursor, subsequently cleaved by the inflammasome to secrete mature IL-1ß. In contrast, neither pro-IL-1ß induction nor mature IL-1ß secretion occurs upon internalization of TiO2 or FePO4 nanoparticles. The pro-IL-1ß induction is suppressed by pharmacologic inhibitors of endosomal TLR activation or by genetic ablation of MyD88, a downstream adapter of TLR pathways, indicating that endosomal pattern recognition is responsible for the observed cytokine response to food-grade SAS particles. CONCLUSIONS: Our results unexpectedly show that food-grade SAS particles are able to directly initiate the endosomal MyD88-dependent pathogen pattern recognition and signaling pathway in steady-state DCs. The ensuing activation of immature DCs with de novo induction of pro-IL-1ß implies that the currently massive use of SAS particles as food additive should be reconsidered.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Aditivos Alimentarios/toxicidad , Interleucina-1beta/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Precursores de Proteínas/metabolismo , Dióxido de Silicio/toxicidad , Animales , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/ultraestructura , Relación Dosis-Respuesta a Droga , Endocitosis , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Endosomas/ultraestructura , Aditivos Alimentarios/síntesis química , Aditivos Alimentarios/metabolismo , Inocuidad de los Alimentos , Inflamasomas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Nanopartículas , Procesamiento Proteico-Postraduccional , Receptores de Reconocimiento de Patrones/metabolismo , Medición de Riesgo , Transducción de Señal/efectos de los fármacos , Dióxido de Silicio/síntesis química , Dióxido de Silicio/metabolismo , Factores de Tiempo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Regulación hacia Arriba
4.
Int J Mol Sci ; 18(2)2017 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-28212334

RESUMEN

Rotaviruses (RVs) are important enteric pathogens of newborn humans and animals, causing diarrhea and in rare cases death, especially in very young individuals. Rotavirus vaccines presently used are modified live vaccines that lack complete biological safety. Previous work from our laboratory suggested that vaccines based on in situ produced, non-infectious rotavirus-like particles (RVLPs) are efficient while being entirely safe. However, using either vaccine, active mucosal immunization cannot induce protective immunity in newborns due to their immature immune system. We therefore hypothesized that offspring from vaccinated dams are passively immunized either by transfer of maternal antibodies during pregnancy or by taking up antibodies from milk. Using a codon optimized polycistronic gene expression cassette packaged into herpesvirus particles, the simultaneous expression of the RV capsid genes led to the intracellular formation of RVLPs in various cell lines. Vaccinated dams developed a strong RV specific IgG antibody response determined in sera and milk of both mother and pups. Moreover, sera of naïve pups nursed by vaccinated dams also had RV specific antibodies suggesting a lactogenic transfer of antibodies. Although full protection of pups was not achieved in this mouse model, our observations are important for the development of improved vaccines against RV in humans as well as in various animal species.


Asunto(s)
Anticuerpos Antivirales/inmunología , Vectores Genéticos/genética , Herpesvirus Humano 1/genética , Leche/inmunología , Vacunas contra Rotavirus/genética , Vacunas contra Rotavirus/inmunología , Rotavirus/inmunología , Vacunación , Animales , Anticuerpos Antivirales/sangre , Especificidad de Anticuerpos , Línea Celular Tumoral , Chlorocebus aethiops , Codón , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Embarazo , Infecciones por Rotavirus/inmunología , Infecciones por Rotavirus/prevención & control , Vacunas contra Rotavirus/administración & dosificación , Transducción Genética , Vacunas de Partículas Similares a Virus/administración & dosificación , Vacunas de Partículas Similares a Virus/genética , Vacunas de Partículas Similares a Virus/inmunología , Células Vero , Proteínas Estructurales Virales/genética , Proteínas Estructurales Virales/inmunología
5.
J Nanobiotechnology ; 14(1): 44, 2016 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-27287345

RESUMEN

The development of nano-materials is viewed as one of the most important technological advances of the 21st century and new applications of nano-sized particles in the production, processing, packaging or storage of food are expected to emerge soon. This trend of growing commercialization of engineered nano-particles as part of modern diet will substantially increase oral exposure. Contrary to the proven benefits of nano-materials, however, possible adverse health effects have generally received less attention. This problem is very well illustrated by nano-structured synthetic amorphous silica (SAS), which is a common food additive since several decades although the relevant risk assessment has never been satisfactorily completed. A no observed adverse effect level of 2500 mg SAS particles/kg body weight per day was derived from the only available long-term administration study in rodents. However, extrapolation to a safe daily intake for humans is problematic due to limitations of this chronic animal study and knowledge gaps as to possible local intestinal effects of SAS particles, primarily on the gut-associated lymphoid system. This uncertainty is aggravated by digestion experiments indicating that dietary SAS particles preserve their nano-sized structure when reaching the intestinal lumen. An important aspect is whether food-borne particles like SAS alter the function of dendritic cells that, embedded in the intestinal mucosa, act as first-line sentinels of foreign materials. We conclude that nano-particles do not represent a completely new threat and that most potential risks can be assessed following procedures established for conventional chemical hazards. However, specific properties of food-borne nano-particles should be further examined and, for that purpose, in vitro tests with decision-making cells of the immune system are needed to complement existing in vivo studies.


Asunto(s)
Aditivos Alimentarios/efectos adversos , Nanoestructuras/efectos adversos , Dióxido de Silicio/efectos adversos , Animales , Aditivos Alimentarios/administración & dosificación , Aditivos Alimentarios/química , Análisis de los Alimentos , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/metabolismo , Humanos , Nanoestructuras/administración & dosificación , Nanoestructuras/química , Medición de Riesgo , Dióxido de Silicio/administración & dosificación , Dióxido de Silicio/química
6.
J Virol ; 88(24): 14396-411, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25297997

RESUMEN

UNLABELLED: Double-stranded RNA (dsRNA) is an important molecular pattern associated with viral infection and is detected by various extra- and intracellular recognition molecules. Poxviruses have evolved to avoid producing dsRNA early in infection but generate significant amounts of dsRNA late in infection due to convergent transcription of late genes. Protein kinase R (PKR) is activated by dsRNA and triggers major cellular defenses against viral infection, including protein synthesis shutdown, apoptosis, and type I interferon (IFN-I) production. The poxviral E3 protein binds and sequesters viral dsRNA and is a major antagonist of the PKR pathway. We found that the highly replication-restricted modified vaccinia virus Ankara (MVA) engineered to produce excess amounts of dsRNA early in infection showed enhanced induction of IFN-ß in murine and human cells in the presence of an intact E3L gene. IFN-ß induction required a minimum overlap length of 300 bp between early complementary transcripts and was strongly PKR dependent. Excess early dsRNA produced by MVA activated PKR early but transiently in murine cells and induced enhanced systemic levels of IFN-α, IFN-γ, and other cytokines and chemokines in mice in a largely PKR-dependent manner. Replication-competent chorioallantois vaccinia virus Ankara (CVA) generating excess early dsRNA also enhanced IFN-I production and was apathogenic in mice even at very high doses but showed no in vitro host range defect. Thus, genetically adjuvanting MVA and CVA to generate excess early dsRNA is an effective method to enhance innate immune stimulation by orthopoxvirus vectors and to attenuate replicating vaccinia virus in vivo. IMPORTANCE: Efficient cellular sensing of pathogen-specific components, including double-stranded RNA (dsRNA), is an important prerequisite of an effective antiviral immune response. The prototype poxvirus vaccinia virus (VACV) and its derivative modified vaccinia virus Ankara (MVA) produce dsRNA as a by-product of viral transcription. We found that inhibition of cellular dsRNA recognition established by the virus-encoded proteins E3 and K3 can be overcome by directing viral overexpression of dsRNA early in infection without compromising replication of MVA in permissive cells. Early dsRNA induced transient activation of the cellular dsRNA sensor protein kinase R (PKR), resulting in enhanced production of interferons and cytokines in cells and mice. Enhancing the capacity of MVA to activate the innate immune system is an important approach to further improve the immunogenicity of this promising vaccine vector.


Asunto(s)
Inmunidad Innata , ARN Bicatenario/inmunología , Virus Vaccinia/inmunología , eIF-2 Quinasa/inmunología , Animales , Línea Celular , Citocinas/metabolismo , Humanos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , ARN Bicatenario/metabolismo , Virus Vaccinia/genética , eIF-2 Quinasa/metabolismo
7.
J Immunol ; 188(8): 3774-83, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22422879

RESUMEN

Plasmacytoid dendritic cells (pDC) are the producers of type I IFNs in response to TLR9 ligands. However, we have found that when bone marrow is depleted of pDC, the IFN-α produced in response to TLR9 ligands is not fully removed. We assign the source of this non-pDC IFN-α as a newly described DC type. It displays the high IFN-α producing activity of pDC but to a more limited range of viruses. Unlike pDC, the novel DC display high T cell stimulation capacity. Moreover, unlike mouse pDC, they are matured with GM-CSF and are less prone to apoptosis upon activation stimuli, including viruses. We propose that these DC constitute a novel bone marrow inflammatory DC type, ideally geared to linking innate and adaptive immune responses in bone marrow via their potent IFN-α production and high T cell stimulatory capacity.


Asunto(s)
Células de la Médula Ósea/inmunología , Células Dendríticas/inmunología , Interferón-alfa/inmunología , Linfocitos T/inmunología , Inmunidad Adaptativa , Animales , Apoptosis , Células de la Médula Ósea/citología , Células Dendríticas/citología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Inmunidad Innata , Interferón-alfa/biosíntesis , Lipopolisacáridos/farmacología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Poli I-C/farmacología , Transducción de Señal , Linfocitos T/citología , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología , Virus Vaccinia/genética , Virus Vaccinia/inmunología
8.
J Virol ; 86(4): 2323-36, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22171261

RESUMEN

Sustained activation of the Raf/MEK/extracellular signal-regulated kinase (ERK) pathway in infected cells has been shown to be crucial for full replication efficiency of orthopoxviruses in cell culture. In infected cells, this pathway is mainly activated by the vaccinia virus growth factor (VGF), an epidermal growth factor (EGF)-like protein. We show here that chorioallantois vaccinia virus Ankara (CVA), but not modified vaccinia virus Ankara (MVA), induced sustained activation of extracellular signal-regulated kinase 1/2 (ERK1/2) in infected human 293 cells, although both viruses direct secretion of functional VGF. A CVA mutant lacking the O1L gene (CVA-ΔO1L) demonstrated that the O1 protein was required for sustained upregulation of the ERK1/2 pathway in 293 cells as well as in other mammalian cell lines. The highly conserved orthopoxvirus O1L gene encodes a predicted 78-kDa protein with a hitherto-unknown function. CVA-ΔO1L showed reduced plaque size and an attenuated cytopathic effect (CPE) in infected cell cultures and reduced virulence and spread from lungs to ovaries in intranasally infected BALB/c mice. Reinsertion of an intact O1L gene into MVA, which in its original form harbors a fragmented O1L open reading frame (ORF), restored ERK1/2 activation in 293 cells but did not increase replication and spread of MVA in human or other mammalian cell lines. Thus, the O1 protein was crucial for sustained ERK1/2 activation in CVA- and MVA-infected human cells, complementing the autocrine function of VGF, and enhanced virulence in vivo.


Asunto(s)
Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Virus Vaccinia/metabolismo , Virus Vaccinia/patogenicidad , Vaccinia/enzimología , Proteínas Virales/metabolismo , Animales , Línea Celular , Activación Enzimática , Femenino , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos BALB C , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Vaccinia/metabolismo , Vaccinia/virología , Virus Vaccinia/genética , Proteínas Virales/genética , Virulencia
9.
Int Rev Cell Mol Biol ; 379: 87-142, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37541728

RESUMEN

Cancer immunotherapy relies on unleashing the patient´s immune system against tumor cells. Cancer vaccines aim to stimulate both the innate and adaptive arms of immunity to achieve durable clinical responses. Some roadblocks for a successful cancer vaccine in the clinic include the tumor antigen of choice, the adjuvants employed to strengthen antitumor-specific immune responses, and the risks associated with enhancing immune-related adverse effects in patients. Modified vaccinia Ankara (MVA) belongs to the family of poxviruses and is a versatile vaccine platform that combines several attributes crucial for cancer therapy. First, MVA is an excellent inducer of innate immune responses leading to type I interferon secretion and induction of T helper cell type 1 (Th1) immune responses. Second, it elicits robust and durable humoral and cellular immunity against vector-encoded heterologous antigens. Third, MVA has enormous genomic flexibility, which allows for the expression of multiple antigenic and costimulatory entities. And fourth, its replication deficit in human cells ensures a excellent safety profile. In this review, we summarize the current understanding of how MVA induces innate and adaptive immune responses. Furthermore, we will give an overview of the tumor-associated antigens and immunomodulatory molecules that have been used to armor MVA and describe their clinical use. Finally, the route of MVA immunization and its impact on therapeutic efficacy depending on the immunomodulatory molecules expressed will be discussed.


Asunto(s)
Neoplasias , Vaccinia , Humanos , Neoplasias/terapia , Virus Vaccinia/genética , Vacunación , Inmunidad Innata
10.
Front Immunol ; 13: 841471, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35774800

RESUMEN

Respiratory syncytial virus (RSV) causes a respiratory disease with a potentially fatal outcome especially in infants and elderly individuals. Several vaccines failed in pivotal clinical trials, and to date, no vaccine against RSV has been licensed. We have developed an RSV vaccine based on the recombinant Modified Vaccinia Virus Ankara-BN® (MVA-RSV), containing five RSV-specific antigens that induced antibody and T-cell responses, which is currently tested in clinical trials. Here, the immunological mechanisms of protection were evaluated to determine viral loads in lungs upon vaccination of mice with MVA-RSV followed by intranasal RSV challenge. Depletion of CD4 or CD8 T cells, serum transfer, and the use of genetically engineered mice lacking the ability to generate either RSV-specific antibodies (T11µMT), the IgA isotype (IgA knockout), or CD8 T cells (ß2M knockout) revealed that complete protection from RSV challenge is dependent on CD4 and CD8 T cells as well as antibodies, including IgA. Thus, MVA-RSV vaccination optimally protects against RSV infection by employing multiple arms of the adaptive immune system.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio , Vacunas contra Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Anciano , Animales , Anticuerpos Antivirales , Formación de Anticuerpos , Humanos , Inmunoglobulina A , Ratones , Virus Vaccinia/genética
11.
J Clin Invest ; 118(5): 1776-84, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18398511

RESUMEN

Poxviruses such as the causative agent of smallpox have developed multiple strategies to suppress immune responses, including the suppression of DC activation. Since poxviruses are large DNA viruses, we hypothesized that their detection by DCs may involve the endosomal DNA recognition receptor TLR9. Indeed, we have shown here that DC recognition of ectromelia virus (ECTV), the causative agent of mousepox, completely depended on TLR9. The importance of TLR9 was highlighted by the fact that mice lacking TLR9 showed drastically increased susceptibility to infection with ECTV. In contrast, we found that the strongly attenuated poxvirus modified vaccinia virus Ankara (MVA) activated DCs by both TLR9-dependent and -independent pathways. We therefore tested whether we could use the broader induction of immune responses by MVA to protect mice from a lethal infection with ECTV. Indeed, MVA given at the same time as a lethal dose of ECTV protected mice from death. Importantly, MVA also rescued TLR9-deficient mice if administered 2 full days after an otherwise lethal infection with ECTV. Therefore, these data suggest an essential role for TLR9 in the defense against poxviruses. In addition, postexposure application of MVA may protect against lethal poxvirus infection.


Asunto(s)
Células Dendríticas/inmunología , Infecciones por Poxviridae , Receptor Toll-Like 9/inmunología , Vacunación , Animales , Células Dendríticas/citología , Virus de la Ectromelia/inmunología , Virus de la Ectromelia/patogenicidad , Humanos , Inmunidad/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Poxviridae/inmunología , Infecciones por Poxviridae/mortalidad , Infecciones por Poxviridae/prevención & control , Tasa de Supervivencia , Receptor Toll-Like 9/genética , Virus Vaccinia/inmunología
12.
J Virol ; 84(17): 8743-52, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20538860

RESUMEN

Efficient T-cell responses against recombinant antigens expressed by vaccinia virus vectors require expression of these antigens in the early phase of the virus replication cycle. The kinetics of recombinant gene expression in poxviruses are largely determined by the promoter chosen. We used the highly attenuated modified vaccinia virus Ankara (MVA) to determine the role of promoters in the induction of CD8 T-cell responses. We constructed MVA recombinants expressing either enhanced green fluorescent protein (EGFP) or chicken ovalbumin (OVA), each under the control of a hybrid early-late promoter (pHyb) containing five copies of a strong early element or the well-known early-late p7.5 or pS promoter for comparison. In primary or cultured cells, EGFP expression under the control of pHyb was detected within 30 min, as an immediate-early protein, and remained higher over the first 6 h of infection than p7.5- or pS-driven EGFP expression. Repeated immunizations of mice with recombinant MVA expressing OVA under the control of the pHyb promoter led to superior acute and memory CD8 T-cell responses compared to those to p7.5- and pS-driven OVA. Moreover, OVA expressed under the control of pHyb replaced the MVA-derived B8R protein as the immunodominant CD8 T-cell antigen after three or more immunizations. This is the first demonstration of an immediate-early neoantigen expressed by a poxviral vector resulting in superior induction of neoantigen-specific CD8 T-cell responses.


Asunto(s)
Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Expresión Génica , Vectores Genéticos/genética , Regiones Promotoras Genéticas , Virus Vaccinia/genética , Vaccinia/inmunología , Animales , Anticuerpos Antivirales/sangre , Antígenos Virales/genética , Secuencia de Bases , Antígenos CD8/genética , Antígenos CD8/inmunología , Linfocitos T CD8-positivos/virología , Línea Celular , Células Cultivadas , Embrión de Pollo , Cricetinae , Femenino , Genes Inmediatos-Precoces , Vectores Genéticos/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Ovalbúmina/genética , Ovalbúmina/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Vaccinia/virología , Virus Vaccinia/química , Virus Vaccinia/inmunología , Vacunas Virales/genética , Vacunas Virales/inmunología
13.
J Virol ; 84(19): 9907-19, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20668072

RESUMEN

Modified vaccinia virus Ankara (MVA) has a highly restricted host range in cell culture and is apathogenic in vivo. MVA was derived from the parental chorioallantois vaccinia virus Ankara (CVA) by more than 570 passages in chicken embryo fibroblast (CEF) cells. During CEF cell passaging, six major deletions comprising 24,668 nucleotides occurred in the CVA genome. We have cloned both the MVA and the parental CVA genome as bacterial artificial chromosomes (BACs) and have sequentially introduced the six major MVA deletions into the cloned CVA genome. Reconstituted mutant CVA viruses containing up to six major MVA deletions showed no detectable replication restriction in 12 of 14 mammalian cell lines tested; the exceptions were rabbit cell lines RK13 and SIRC. In mice, CVA mutants with up to three deletions showed slightly enhanced virulence, suggesting that gene deletion in replicating vaccinia virus (VACV) can result in gain of fitness in vivo. CVA mutants containing five or all six deletions were still pathogenic, with a moderate degree of attenuation. Deletion V was mainly responsible for the attenuated phenotype of these mutants. In conclusion, loss or truncation of all 31 open reading frames in the six major deletions is not sufficient to reproduce the specific MVA phenotype of strong attenuation and highly restricted host range. Mutations in viral genes outside or in association with the six major deletions appear to contribute significantly to this phenotype. Host range restriction and avirulence of MVA are most likely a cooperative effect of gene deletions and mutations involving the major deletions.


Asunto(s)
Eliminación de Gen , Genoma Viral , Virus Vaccinia/genética , Virus Vaccinia/patogenicidad , Animales , Línea Celular , Embrión de Pollo , Cromosomas Artificiales Bacterianos/genética , Efecto Citopatogénico Viral , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Fenotipo , Conejos , Recombinación Genética , Vaccinia/etiología , Vaccinia/virología , Virus Vaccinia/fisiología , Virulencia/genética , Cultivo de Virus , Replicación Viral
14.
Int Immunol ; 22(9): 749-56, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20584765

RESUMEN

Arenaviruses such as Lassa virus (LASV) cause significant morbidity and mortality in endemic areas. Using a glycoprotein (GP) exchange strategy, we have recently developed live-attenuated arenavirus vaccine prototypes (rLCMV/VSVG) based on lymphocytic choriomeningitis virus (LCMV), a close relative of LASV. rLCMV/VSVG induced long-term CD8(+) T cell immunity against wild-type virus challenge and exhibited a stably attenuated phenotype in vivo. Here we elucidated the innate and adaptive immune requirements for the control of rLCMV/VSVG. Infection of RAG(-/-) mice resulted in persisting viral RNA in blood but not in overt viremia. The latter was only found in mice lacking both RAG and IFN type I receptor. Conversely, absence of IFN type II signaling or NK cells on an RAG-deficient background had only minor effects on vaccine virus load or none at all. rLCMV/VSVG infection of wild-type mice induced less type I IFN than did wild-type LCMV, and type I as well as type II IFNs were dispensable for the induction of virus-specific memory CD8 T cells and virus-neutralizing antibodies by rLCMV/VSVG. In conclusion, the adaptive immune systems are essential for elimination of rLCMV/VSVG, and type I but not type II IFN plays a major contributive role in lowering rLCMV/VSVG loads in vivo, attesting to the attenuation profile of the vaccine. Nevertheless, IFNs are not required for the induction of potent vaccine responses. These results provide a better understanding of the immunobiology of rLCMV/VSVG and will contribute to the further development of GP exchange vaccines for combating arenaviral hemorrhagic fevers.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Virus Lassa/inmunología , Receptor de Interferón alfa y beta/metabolismo , Receptores de Interferón/metabolismo , Vacunas Virales , Inmunidad Adaptativa , Animales , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Genes RAG-1/genética , Inmunidad Innata , Memoria Inmunológica , Fiebre de Lassa/inmunología , Fiebre de Lassa/prevención & control , Virus Lassa/patogenicidad , Ratones , Ratones Endogámicos C57BL , Organismos Modificados Genéticamente , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Vacunas Atenuadas , Receptor de Interferón gamma
15.
J Immunother Cancer ; 9(2)2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33579736

RESUMEN

Background Human cancers are extraordinarily heterogeneous in terms of tumor antigen expression, immune infiltration and composition. A common feature, however, is the host's inability to mount potent immune responses that prevent tumor growth effectively. Often, naturally primed CD8+ T cells against solid tumors lack adequate stimulation and efficient tumor tissue penetration due to an immune hostile tumor microenvironment.Methods To address these shortcomings, we cloned tumor-associated antigens (TAA) and the immune-stimulatory ligand 4-1BBL into the genome of modified vaccinia Ankara (MVA) for intratumoral virotherapy.Results Local treatment with MVA-TAA-4-1BBL resulted in control of established tumors. Intratumoral injection of MVA localized mainly to the tumor with minimal leakage to the tumor-draining lymph node. In situ infection by MVA-TAA-4-1BBL triggered profound changes in the tumor microenvironment, including the induction of multiple proinflammatory molecules and immunogenic cell death. These changes led to the reactivation and expansion of antigen-experienced, tumor-specific cytotoxic CD8+ T cells that were essential for the therapeutic antitumor effect. Strikingly, we report the induction of a systemic antitumor immune response including tumor antigen spread by local MVA-TAA-4-1BBL treatment which controlled tumor growth at distant, untreated lesions and protected against local and systemic tumor rechallenge. In all cases, 4-1BBL adjuvanted MVA was superior to MVA.Conclusion Intratumoral 4-1BBL-armed MVA immunotherapy induced a profound reactivation and expansion of potent tumor-specific CD8+ T cells as well as favorable proinflammatory changes in the tumor microenvironment, leading to elimination of tumors and protective immunological memory.


Asunto(s)
Ligando 4-1BB/genética , Antígenos de Neoplasias/genética , Melanoma Experimental/terapia , Viroterapia Oncolítica/métodos , Virus Vaccinia/fisiología , Ligando 4-1BB/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Clonación Molecular , Terapia Combinada , Sinergismo Farmacológico , Femenino , Memoria Inmunológica , Melanoma Experimental/inmunología , Ratones , Resultado del Tratamiento , Microambiente Tumoral , Virus Vaccinia/genética
16.
J Exp Med ; 199(5): 731-6, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-14981113

RESUMEN

Psoriasis is a common T cell-mediated autoimmune disorder where primary onset of skin lesions is followed by chronic relapses. Progress in defining the mechanism for initiation of pathological events has been hampered by the lack of a relevant experimental model in which psoriasis develops spontaneously. We present a new animal model in which skin lesions spontaneously developed when symptomless prepsoriatic human skin was engrafted onto AGR129 mice, deficient in type I and type II interferon receptors and for the recombination activating gene 2. Upon engraftment, resident human T cells in prepsoriatic skin underwent local proliferation. T cell proliferation was crucial for development of a psoriatic phenotype because blocking of T cells led to inhibition of psoriasis development. Tumor necrosis factor-alpha was a key regulator of local T cell proliferation and subsequent disease development. Our observations highlight the importance of resident T cells in the context of lesional tumor necrosis factor-alpha production during development of a psoriatic lesion. These findings underline the importance of resident immune cells in psoriasis and will have implications for new therapeutic strategies for psoriasis and other T cell-mediated diseases.


Asunto(s)
Psoriasis/etiología , Psoriasis/inmunología , Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , División Celular , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Modelos Animales de Enfermedad , Humanos , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares , Fenotipo , Psoriasis/patología , Receptor de Interferón alfa y beta , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Trasplante de Piel , Linfocitos T/patología , Trasplante Heterólogo
17.
J Exp Med ; 197(5): 575-84, 2003 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-12615899

RESUMEN

Flt-3 ligand (FL), a hematopoetic growth factor, increases the number of dendritic cells (DCs), B cells, and natural killer cells in adult mice but the effect in neonates was unknown. We show that FL treatment of newborn mice induced a >100-fold increase in the innate resistance against infection with herpes simplex virus type 1 and Listeria monocytogenes. This resistance required interferon (IFN)-alpha/beta for viral and interleukin (IL)-12 for bacterial infections. Long-term survival after viral but not bacterial infection was increased approximately 100-fold by FL treatment. After treatment, CD11c(+)/major histocompatibility complex type II(+) and CD11c(+)/B220(+) DC lineage cells were the only cell populations increased in the spleen, liver, peritoneum, and skin. DC induction was independent of IFNs, IL-2, -4, -7, -9, -15, and mature T and B cells. The data suggest that FL increases the number of DCs in neonates and possibly in other immune-compromised individuals, which in turn improves IFN-alpha/beta- and IL-12-associated immune responses.


Asunto(s)
Herpes Simple/inmunología , Herpesvirus Humano 1 , Inmunidad Innata , Listeria monocytogenes , Listeriosis/inmunología , Proteínas de la Membrana/farmacología , Animales , Animales Recién Nacidos , Linfocitos B/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Citometría de Flujo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Interferón-alfa/metabolismo , Interferón beta/metabolismo , Interleucina-12/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos , Ratones Noqueados , Piel/citología , Piel/metabolismo , Bazo/citología , Bazo/metabolismo , Tasa de Supervivencia
18.
Eur J Immunol ; 39(1): 26-35, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19137537

RESUMEN

Newborns face complex physical and immunological changes before and after birth. Although the uterus is a sterile environment for the fetus, it also contains non-self material from the mother. Birth involves the transition from the sterile intra-uterine environment to an environment rich in microbes and requires rapid induction of appropriate responses to control these microbes. In this review we focus on the similarities and differences of human and murine neonatal DC and their reaction to various stimuli. A better understanding of the newborn immune system--in particular, the DC-T-cell interaction--will be beneficial for the development of improved strategies to prevent or treat infections in this vulnerable population and prepare the immune system to cope with allergens and tumors later in life.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Células Dendríticas/inmunología , Sistema Inmunológico/inmunología , Recién Nacido/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Biomarcadores , Citocinas/inmunología , Humanos , Proteínas de la Membrana/inmunología , Receptores Toll-Like/inmunología
19.
Nat Commun ; 10(1): 5041, 2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31695037

RESUMEN

Virus-based vaccines and appropriate costimulation potently enhance antigen-specific T cell immunity against cancer. Here we report the use of recombinant modified vaccinia virus Ankara (rMVA) encoding costimulatory CD40L against solid tumors. Therapeutic treatment with rMVA-CD40L-expressing tumor-associated antigens results in the control of established tumors. The expansion of tumor-specific cytotoxic CD8+ T cells is essential for the therapeutic antitumor effects. Strikingly, rMVA-CD40L also induces strong natural killer (NK) cell activation and expansion. Moreover, the combination of rMVA-CD40L and tumor-targeting antibodies results in increased therapeutic antitumor efficacy relying on the presence of Fc receptor and NK cells. We describe a translationally relevant therapeutic synergy between systemic viral vaccination and CD40L costimulation. We show strengthened antitumor immune responses when both rMVA-CD40L-induced innate and adaptive immune mechanisms are exploited by combination with tumor-targeting antibodies. This immunotherapeutic approach could translate into clinical cancer therapies where tumor-targeting antibodies are employed.


Asunto(s)
Inmunidad Adaptativa , Anticuerpos Antineoplásicos/inmunología , Ligando de CD40/farmacología , Vacunas contra el Cáncer/inmunología , Inmunidad Innata , Inmunoterapia/métodos , Neoplasias/terapia , Vacunas Virales/uso terapéutico , Adyuvantes Inmunológicos/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Sinergismo Farmacológico , Femenino , Humanos , Inmunización , Células Asesinas Naturales/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias/inmunología , Vacunación , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA