Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Nature ; 592(7853): 283-289, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33524990

RESUMO

A safe and effective vaccine against COVID-19 is urgently needed in quantities that are sufficient to immunize large populations. Here we report the preclinical development of two vaccine candidates (BNT162b1 and BNT162b2) that contain nucleoside-modified messenger RNA that encodes immunogens derived from the spike glycoprotein (S) of SARS-CoV-2, formulated in lipid nanoparticles. BNT162b1 encodes a soluble, secreted trimerized receptor-binding domain (known as the RBD-foldon). BNT162b2 encodes the full-length transmembrane S glycoprotein, locked in its prefusion conformation by the substitution of two residues with proline (S(K986P/V987P); hereafter, S(P2) (also known as P2 S)). The flexibly tethered RBDs of the RBD-foldon bind to human ACE2 with high avidity. Approximately 20% of the S(P2) trimers are in the two-RBD 'down', one-RBD 'up' state. In mice, one intramuscular dose of either candidate vaccine elicits a dose-dependent antibody response with high virus-entry inhibition titres and strong T-helper-1 CD4+ and IFNγ+CD8+ T cell responses. Prime-boost vaccination of rhesus macaques (Macaca mulatta) with the BNT162b candidates elicits SARS-CoV-2-neutralizing geometric mean titres that are 8.2-18.2× that of a panel of SARS-CoV-2-convalescent human sera. The vaccine candidates protect macaques against challenge with SARS-CoV-2; in particular, BNT162b2 protects the lower respiratory tract against the presence of viral RNA and shows no evidence of disease enhancement. Both candidates are being evaluated in phase I trials in Germany and the USA1-3, and BNT162b2 is being evaluated in an ongoing global phase II/III trial (NCT04380701 and NCT04368728).


Assuntos
Vacinas contra COVID-19/imunologia , COVID-19/imunologia , COVID-19/prevenção & controle , Modelos Animais de Doenças , SARS-CoV-2/imunologia , Envelhecimento/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Antígenos Virais/química , Antígenos Virais/genética , Antígenos Virais/imunologia , Vacina BNT162 , COVID-19/sangue , COVID-19/terapia , COVID-19/virologia , Vacinas contra COVID-19/administração & dosagem , Vacinas contra COVID-19/química , Vacinas contra COVID-19/genética , Linhagem Celular , Ensaios Clínicos como Assunto , Feminino , Humanos , Imunização Passiva , Internacionalidade , Macaca mulatta/imunologia , Macaca mulatta/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Multimerização Proteica , RNA Viral/análise , Sistema Respiratório/imunologia , Sistema Respiratório/virologia , SARS-CoV-2/química , SARS-CoV-2/genética , Solubilidade , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia , Linfócitos T/imunologia , Vacinação , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/química , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Soroterapia para COVID-19 , Vacinas de mRNA
2.
Mol Ther ; 31(2): 374-386, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36303436

RESUMO

Emerging and re-emerging viruses, such as Zaire Ebola virus (EBOV), pose a global threat and require immediate countermeasures, including the rapid development of effective vaccines that are easy to manufacture. Synthetic self-amplifying RNAs (saRNAs) attend to these needs, being safe and strong immune stimulators that can be inexpensively produced in large quantities, using cell-free systems and good manufacturing practice. Here, the first goal was to develop and optimize an anti-EBOV saRNA-based vaccine in terms of its antigen composition and route of administration. Vaccinating mice with saRNAs expressing the EBOV glycoprotein (GP) alone or in combination with the nucleoprotein (NP) elicited antigen-specific immune responses. GP-specific antibodies showed neutralizing activity against EBOV. Strong CD4+ T cell response against NP and GP and CD8+ T cell response against NP were detected by ELISpot assays. Intramuscular vaccination with saRNAs conferred better immune response than intradermal. Finally, mice vaccinated in a prime-boost regimen with saRNAs encoding both GP and NP or with GP alone survived an EBOV infection. In addition, a single dose of GP and NP saRNAs was also protective against fatal EBOV infection. Overall, saRNAs expressing viral antigens represent a promising vaccine platform.


Assuntos
Vacinas contra Ebola , Ebolavirus , Doença pelo Vírus Ebola , Animais , Camundongos , Doença pelo Vírus Ebola/prevenção & controle , Anticorpos Antivirais , Anticorpos Neutralizantes , Ebolavirus/genética , Glicoproteínas/genética , Vacinas contra Ebola/genética
3.
Mol Ther ; 28(1): 119-128, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31624015

RESUMO

Here, we present a potent RNA vaccine approach based on a novel bipartite vector system using trans-amplifying RNA (taRNA). The vector cassette encoding the vaccine antigen originates from an alphaviral self-amplifying RNA (saRNA), from which the replicase was deleted to form a transreplicon. Replicase activity is provided in trans by a second molecule, either by a standard saRNA or an optimized non-replicating mRNA (nrRNA). The latter delivered 10- to 100-fold higher transreplicon expression than the former. Moreover, expression driven by the nrRNA-encoded replicase in the taRNA system was as efficient as in a conventional monopartite saRNA system. We show that the superiority of nrRNA- over saRNA-encoded replicase to drive expression of the transreplicon is most likely attributable to its higher translational efficiency and lack of interference with cellular translation. Testing the novel taRNA system in mice, we observed that doses of influenza hemagglutinin antigen-encoding RNA as low as 50 ng were sufficient to induce neutralizing antibodies and mount a protective immune response against live virus challenge. These findings, together with a favorable safety profile, a simpler production process, and the universal applicability associated with this bipartite vector system, warrant further exploration of taRNA.


Assuntos
Imunogenicidade da Vacina , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/administração & dosagem , Influenza Humana/metabolismo , Infecções por Orthomyxoviridae/prevenção & controle , RNA Viral/genética , Vírus da Floresta de Semliki/genética , Vacinação , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Cricetinae , Cães , Feminino , Vetores Genéticos , Células HEK293 , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Vacinas contra Influenza/imunologia , Influenza Humana/virologia , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/virologia , Proteínas do Complexo da Replicase Viral/genética
4.
Mol Ther ; 26(2): 446-455, 2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29275847

RESUMO

New vaccine platforms are needed to address the time gap between pathogen emergence and vaccine licensure. RNA-based vaccines are an attractive candidate for this role: they are safe, are produced cell free, and can be rapidly generated in response to pathogen emergence. Two RNA vaccine platforms are available: synthetic mRNA molecules encoding only the antigen of interest and self-amplifying RNA (sa-RNA). sa-RNA is virally derived and encodes both the antigen of interest and proteins enabling RNA vaccine replication. Both platforms have been shown to induce an immune response, but it is not clear which approach is optimal. In the current studies, we compared synthetic mRNA and sa-RNA expressing influenza virus hemagglutinin. Both platforms were protective, but equivalent levels of protection were achieved using 1.25 µg sa-RNA compared to 80 µg mRNA (64-fold less material). Having determined that sa-RNA was more effective than mRNA, we tested hemagglutinin from three strains of influenza H1N1, H3N2 (X31), and B (Massachusetts) as sa-RNA vaccines, and all protected against challenge infection. When sa-RNA was combined in a trivalent formulation, it protected against sequential H1N1 and H3N2 challenges. From this we conclude that sa-RNA is a promising platform for vaccines against viral diseases.


Assuntos
Vírus da Influenza A/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , RNA Viral/imunologia , Animais , Modelos Animais de Doenças , Feminino , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Imunização , Imunização Secundária , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A/genética , Vacinas contra Influenza/genética , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/imunologia , RNA Viral/genética , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
5.
J Gen Virol ; 97(7): 1511-1519, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27075405

RESUMO

Highly pathogenic Nipah virus (NiV) causes symptomatic infections in pigs and humans. The severity of respiratory symptoms is much more pronounced in pigs than in humans, suggesting species-specific differences of NiV replication in porcine and human airways. Here, we present a comparative study on productive NiV replication in primary airway epithelial cell cultures of the two species. We reveal that NiV growth substantially differs in primary cells between pigs and humans, with a more rapid spread of infection in human airway epithelia. Increased replication, correlated with higher endogenous expression levels of the main NiV entry receptor ephrin-B2, not only significantly differed between airway cells of the two species but also varied between cells from different human donors. To our knowledge, our study provides the first experimental evidence of species-specific and individual differences in NiV receptor expression and replication kinetics in primary airway epithelial cells. It remains to be determined whether and how these differences contribute to the viral host range and pathogenicity.


Assuntos
Efrina-B2/metabolismo , Células Epiteliais/virologia , Infecções por Henipavirus/transmissão , Vírus Nipah/fisiologia , Receptores Virais/metabolismo , Mucosa Respiratória/virologia , Replicação Viral/fisiologia , Animais , Células Cultivadas , Infecções por Henipavirus/virologia , Especificidade de Hospedeiro , Humanos , Vírus Nipah/patogenicidade , Mucosa Respiratória/citologia , Especificidade da Espécie , Suínos , Doenças dos Suínos/virologia , Internalização do Vírus
6.
J Virol ; 87(6): 3143-54, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23283941

RESUMO

Highly pathogenic Nipah virus (NiV) infections are transmitted via airway secretions and urine, commonly via the respiratory route. Epithelial surfaces represent important replication sites in both primary and systemic infection phases. NiV entry and spread from polarized epithelial cells therefore determine virus entry and dissemination within a new host and influence virus shedding via mucosal surfaces in the respiratory and urinary tract. To date, there is no knowledge regarding the entry and exit sites of NiV in polarized epithelial cells. In this report, we show for the first time that NiV can infect polarized kidney epithelial cells (MDCK) from both cell surfaces, while virus release is primarily restricted to the apical plasma membrane. Substantial amounts of basolateral infectivity were detected only after infection with high virus doses, at time points when the integrity of the cell monolayer was largely disrupted as a result of cell-to-cell fusion. Confocal immunofluorescence analyses of envelope protein distribution at early and late infection stages suggested that apical virus budding is determined by the polarized sorting of the NiV matrix protein, M. Studies with stably M-expressing and with monensin-treated cells furthermore demonstrated that M protein transport is independent from the glycoproteins, implying that the M protein possesses an intrinsic apical targeting signal.


Assuntos
Células Epiteliais/virologia , Vírus Nipah/fisiologia , Internalização do Vírus , Liberação de Vírus , Linhagem Celular , Humanos , Microscopia Confocal , Microscopia de Fluorescência , Transporte Proteico , Proteínas da Matriz Viral/metabolismo
7.
Methods Mol Biol ; 2786: 135-144, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38814392

RESUMO

The recent COVID-19 pandemic as well as other past and recent outbreaks of newly or re-emerging viruses show the urgent need to develop potent new vaccine approaches, that enable a quick response to prevent global spread of infectious diseases. The breakthrough of first messenger RNA (mRNA)-based vaccines 2019 approved only months after identification of the causative virus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), opens a big new field for vaccine engineering. Currently, two major types of mRNA are being pursued as vaccines for the prevention of infectious diseases. One is non-replicating mRNA, including nucleoside-modified mRNA, used in the current COVID-19 vaccines of Moderna and BioNTech (Sahin et al., Nat Rev Drug Discov 13(10):759-780, 2014; Baden et al., N Engl J Med 384(5):403-416, 2021; Polack et al., N Engl J Med 383(27):2603-2615, 2020), the other is self-amplifying RNA (saRNA) derived from RNA viruses. Recently, trans-amplifying RNA, a split vector system, has been described as a third class of mRNA (Spuul et al., J Virol 85(10):4739-4751, 2011; Blakney et al., Front Mol Biosci 5:71, 2018; Beissert et al., Mol Ther 28(1):119-128, 2020). In this chapter we review the different types of mRNA currently used for vaccine development with focus on trans-amplifying RNA.


Assuntos
Vacinas contra COVID-19 , COVID-19 , SARS-CoV-2 , Vacinas de mRNA , Humanos , SARS-CoV-2/genética , SARS-CoV-2/imunologia , Vacinas contra COVID-19/imunologia , Vacinas contra COVID-19/genética , COVID-19/prevenção & controle , COVID-19/virologia , COVID-19/imunologia , RNA Viral/genética , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Desenvolvimento de Vacinas , Animais
8.
J Virol ; 86(7): 3736-45, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22278224

RESUMO

Proteolytic activation of the fusion protein of the highly pathogenic Nipah virus (NiV F) is a prerequisite for the production of infectious particles and for virus spread via cell-to-cell fusion. Unlike other paramyxoviral fusion proteins, functional NiV F activation requires endocytosis and pH-dependent cleavage at a monobasic cleavage site by endosomal proteases. Using prototype Vero cells, cathepsin L was previously identified to be a cleavage enzyme. Compared to Vero cells, MDCK cells showed substantially higher F cleavage rates in both NiV-infected and NiV F-transfected cells. Surprisingly, this could not be explained either by an increased F endocytosis rate or by elevated cathepsin L activities. On the contrary, MDCK cells did not display any detectable cathepsin L activity. Though we could confirm cathepsin L to be responsible for F activation in Vero cells, inhibitor studies revealed that in MDCK cells, cathepsin B was required for F-protein cleavage and productive replication of pathogenic NiV. Supporting the idea of an efficient F cleavage in early and recycling endosomes of MDCK cells, endocytosed F proteins and cathepsin B colocalized markedly with the endosomal marker proteins early endosomal antigen 1 (EEA-1), Rab4, and Rab11, while NiV F trafficking through late endosomal compartments was not needed for F activation. In summary, this study shows for the first time that endosomal cathepsin B can play a functional role in the activation of highly pathogenic NiV.


Assuntos
Catepsina B/metabolismo , Endossomos/enzimologia , Infecções por Henipavirus/enzimologia , Infecções por Henipavirus/virologia , Vírus Nipah/metabolismo , Proteínas Virais de Fusão/metabolismo , Animais , Catepsina B/genética , Catepsina L/genética , Catepsina L/metabolismo , Linhagem Celular , Cães , Endocitose , Endossomos/virologia , Infecções por Henipavirus/genética , Infecções por Henipavirus/fisiopatologia , Humanos , Camundongos , Camundongos Knockout , Vírus Nipah/genética , Proteínas Virais de Fusão/genética
9.
Mol Ther Nucleic Acids ; 34: 102045, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37876532

RESUMO

The response to mRNA vaccines needs to be sufficient for immune cell activation and recruitment, but moderate enough to ensure efficacious antigen expression. The choice of the cap structure and use of N1-methylpseudouridine (m1Ψ) instead of uridine, which have been shown to reduce RNA sensing by the cellular innate immune system, has led to improved efficacy of mRNA vaccine platforms. Understanding how RNA modifications influence the cell intrinsic immune response may help in the development of more effective mRNA vaccines. In the current study, we compared mRNA vaccines in mice against influenza virus using three different mRNA formats: uridine-containing mRNA (D1-uRNA), m1Ψ-modified mRNA (D1-modRNA), and D1-modRNA with a cap1 structure (cC1-modRNA). D1-uRNA vaccine induced a significantly different gene expression profile to the modified mRNA vaccines, with an up-regulation of Stat1 and RnaseL, and increased systemic inflammation. This result correlated with significantly reduced antigen-specific antibody responses and reduced protection against influenza virus infection compared with D1-modRNA and cC1-modRNA. Incorporation of m1Ψ alone without cap1 improved antibodies, but both modifications were required for the optimum response. Therefore, the incorporation of m1Ψ and cap1 alters protective immunity from mRNA vaccines by altering the innate immune response to the vaccine material.

10.
J Virol ; 84(15): 7634-41, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20484517

RESUMO

The highly pathogenic Nipah virus (NiV) is aerially transmitted and causes a systemic infection after entering the respiratory tract. Airway epithelia are thus important targets in primary infection. Furthermore, virus replication in the mucosal surfaces of the respiratory or urinary tract in later phases of infection is essential for virus shedding and transmission. So far, the mechanisms of NiV replication in epithelial cells are poorly elucidated. In the present study, we provide evidence that bipolar targeting of the two NiV surface glycoproteins G and F is of biological importance for fusion in polarized epithelia. We demonstrate that infection of polarized cells induces focus formation, with both glycoproteins located at lateral membranes of infected cells adjacent to uninfected cells. Supporting the idea of a direct spread of infection via lateral cell-to-cell fusion, we could identify basolateral targeting signals in the cytoplasmic domains of both NiV glycoproteins. Tyrosine 525 in the F protein is part of an endocytosis signal and is also responsible for basolateral sorting. Surprisingly, we identified a dityrosine motif at position 28/29 in the G protein, which mediates polarized targeting. A dileucine motif predicted to function as sorting signal is not involved. Mutation of the targeting signal in one of the NiV glycoproteins prevented the fusion of polarized cells, suggesting that basolateral or bipolar F and G expression facilitates the spread of NiV within epithelial cell monolayers, thereby contributing to efficient virus spread in mucosal surfaces in early and late phases of infection.


Assuntos
Células Epiteliais/virologia , Vírus Nipah/fisiologia , Tirosina/metabolismo , Proteínas do Envelope Viral/fisiologia , Internalização do Vírus , Replicação Viral , Motivos de Aminoácidos , Animais , Fusão Celular , Endocitose , Humanos
11.
Virol J ; 7: 305, 2010 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-21054904

RESUMO

BACKGROUND: The highly pathogenic Nipah virus (NiV) causes fatal respiratory and brain infections in animals and humans. The major hallmark of the infection is a systemic endothelial infection, predominantly in the CNS. Infection of brain endothelial cells allows the virus to overcome the blood-brain-barrier (BBB) and to subsequently infect the brain parenchyma. However, the mechanisms of NiV replication in endothelial cells are poorly elucidated. We have shown recently that the bipolar or basolateral expression of the NiV surface glycoproteins F and G in polarized epithelial cell layers is involved in lateral virus spread via cell-to-cell fusion and that correct sorting depends on tyrosine-dependent targeting signals in the cytoplasmic tails of the glycoproteins. Since endothelial cells share many characteristics with epithelial cells in terms of polarization and protein sorting, we wanted to elucidate the role of the NiV glycoprotein targeting signals in endothelial cells. RESULTS: As observed in vivo, NiV infection of endothelial cells induced syncytia formation. The further finding that infection increased the transendothelial permeability supports the idea of spread of infection via cell-to-cell fusion and endothelial cell damage as a mechanism to overcome the BBB. We then revealed that both glycoproteins are expressed at lateral cell junctions (bipolar), not only in NiV-infected primary endothelial cells but also upon stable expression in immortalized endothelial cells. Interestingly, mutation of tyrosines 525 and 542/543 in the cytoplasmic tail of the F protein led to an apical redistribution of the protein in endothelial cells whereas tyrosine mutations in the G protein had no effect at all. This fully contrasts the previous results in epithelial cells where tyrosine 525 in the F, and tyrosines 28/29 in the G protein were required for correct targeting. CONCLUSION: We conclude that the NiV glycoprotein distribution is responsible for lateral virus spread in both, epithelial and endothelial cell monolayers. However, the prerequisites for correct protein targeting differ markedly in the two polarized cell types.


Assuntos
Células Endoteliais/virologia , Glicoproteínas/metabolismo , Vírus Nipah/fisiologia , Proteínas do Envelope Viral/metabolismo , Animais , Permeabilidade Capilar , Fusão Celular , Células Cultivadas , Células Gigantes , Suínos
12.
Mol Ther Methods Clin Dev ; 12: 32-46, 2019 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-30547051

RESUMO

Focusing T cell responses on the most vulnerable parts of HIV-1, the functionally conserved regions of HIV-1 proteins, is likely a key prerequisite for vaccine success. For a T cell vaccine to efficiently control HIV-1 replication, the vaccine-elicited individual CD8+ T cells and as a population have to display a number of critical traits. If any one of these traits is suboptimal, the vaccine is likely to fail. Fine-tuning of individual protective characteristics of T cells will require iterative stepwise improvements in clinical trials. Although the second-generation tHIVconsvX immunogens direct CD8+ T cells to predominantly protective and conserved epitopes, in the present work, we have used formulated self-amplifying mRNA (saRNA) to deliver tHIVconsvX to the immune system. We demonstrated in BALB/c and outbred mice that regimens employing saRNA vaccines induced broadly specific, plurifunctional CD8+ and CD4+ T cells, which displayed structured memory subpopulations and were maintained at relatively high frequencies over at least 22 weeks post-administration. This is one of the first thorough analyses of mRNA vaccine-elicited T cell responses. The combination of tHIVconsvX immunogens and the highly versatile and easily manufacturable saRNA platform may provide a long-awaited opportunity to define and optimize induction of truly protective CD8+ T cell parameters in human volunteers.

13.
Virol J ; 5: 142, 2008 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-19036148

RESUMO

Nipah virus (NiV) is a highly pathogenic paramyxovirus that causes severe diseases in animals and humans. Endothelial cell (EC) infection is an established hallmark of NiV infection in vivo. Despite systemic virus spread via the vascular system, EC in brain and lung are preferentially infected whereas EC in other organs are less affected. As in vivo, we found differences in the infection of EC in cell culture. Only brain-derived primary or immortalized EC were found to be permissive to NiV infection. Using a replication-independent fusion assay, we could show that the lack of infection in non-brain EC was due to a lack of receptor expression. The NiV entry receptors ephrinB2 (EB2) or ephrinB3 were only expressed in brain endothelia. The finding that EB2 expression in previously non-permissive aortic EC rendered the cells permissive to infection then demonstrated that EB2 is not only necessary but also sufficient to allow the establishment of a productive NiV infection. This strongly suggests that limitations in receptor expression restrict virus entry in certain EC subsets in vivo, and are thus responsible for the differences in EC tropism observed in human and animal NiV infections.


Assuntos
Efrina-B2/metabolismo , Infecções por Henipavirus/metabolismo , Vírus Nipah/fisiologia , Receptores Virais/metabolismo , Internalização do Vírus , Animais , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Humanos
14.
Virol J ; 5: 163, 2008 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-19108727

RESUMO

BACKGROUND: Cell entry and cell-to-cell spread of the highly pathogenic Nipah virus (NiV) requires binding of the NiV G protein to cellular ephrin receptors and subsequent NiV F-mediated fusion. Since expression levels of the main NiV entry receptor ephrin-B2 (EB2) are highly regulated in vivo to fulfill the physiological functions in axon guidance and angiogenesis, the goal of this study was to determine if changes in the EB2 expression influence NiV infection. RESULTS: Surprisingly, transfection of increasing EB2 plasmid concentrations reduced cell-to-cell fusion both in cells expressing the NiV glycoproteins and in cells infected with NiV. This effect was attributed to the downregulation of the NiV glycoproteins from the cell surface. In addition to the influence on cell-to-cell fusion, increased EB2 expression significantly reduced the total amount of NiV-infected cells, thus interfered with virus entry. To determine if the negative effect of elevated EB2 expression on virus entry is a result of an increased EB2 signaling, receptor function of a tail-truncated and therefore signaling-defective DeltacEB2 was tested. Interestingly, DeltacEB2 fully functioned as NiV entry and fusion receptor, and overexpression also interfered with virus replication. CONCLUSION: Our findings clearly show that EB2 signaling does not account for the striking negative impact of elevated receptor expression on NiV infection, but rather that the ratio between the NiV envelope glycoproteins and surface receptors critically influence cell-to-cell fusion and virus entry.


Assuntos
Efrina-B2/biossíntese , Infecções por Henipavirus/metabolismo , Vírus Nipah/fisiologia , Receptores Virais/biossíntese , Internalização do Vírus , Animais , Chlorocebus aethiops , Citoplasma/metabolismo , Regulação para Baixo , Efrina-B2/genética , Células HeLa , Infecções por Henipavirus/virologia , Humanos , Vírus Nipah/metabolismo , Receptores Virais/genética , Células Vero , Proteínas do Envelope Viral/metabolismo , Ligação Viral
15.
Hum Gene Ther ; 28(12): 1138-1146, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28877647

RESUMO

Among nucleic acid-based delivery platforms, self-amplifying RNA (saRNA) vectors are of increasing interest for applications such as transient expression of recombinant proteins and vaccination. saRNA is safe and, due to its capability to amplify intracellularly, high protein levels can be produced from even minute amounts of transfected templates. However, it is an obstacle to full exploitation of this platform that saRNA induces a strong innate host immune response. In transfected cells, pattern recognition receptors sense double-stranded RNA intermediates and via activation of protein kinase R (PKR) and interferon signaling initiate host defense measures including a translational shutdown. To reduce pattern recognition receptor stimulation and unleash suppressed saRNA translation, this study co-delivered non-replicating mRNA encoding vaccinia virus immune evasion proteins E3, K3, and B18. It was shown that E3 is far superior to K3 or B18 as a highly potent blocker of PKR activation and of interferon (IFN)-ß upregulation. B18, in contrast, is superior in controlling OAS1, a key IFN-inducible gene involved in viral RNA degradation. By combining all three vaccinia proteins, the study achieved significant suppression of PKR and IFN pathway activation in vitro and enhanced expression of saRNA-encoded genes of interest both in vitro and in vivo. This approach promises to overcome key hurdles of saRNA gene delivery. Its application may improve the bioavailability of the encoded protein, and reduce the effective dose and correspondingly the cost of goods of manufacture in the various fields where saRNA utilization is envisioned.


Assuntos
Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Evasão da Resposta Imune , RNA , Vaccinia virus/genética , Proteínas Virais , Animais , Linhagem Celular , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , RNA/genética , RNA/metabolismo , Proteínas Virais/biossíntese , Proteínas Virais/genética , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
16.
Sci Rep ; 7(1): 16892, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29203786

RESUMO

Recombinant vaccine strain-derived measles virus (MV) is clinically tested both as vaccine platform to protect against other pathogens and as oncolytic virus for tumor treatment. To investigate the potential synergism in anti-tumoral efficacy of oncolytic and vaccine properties, we chose Ovalbumin and an ideal tumor antigen, claudin-6, for pre-clinical proof of concept. To enhance immunogenicity, both antigens were presented by retroviral virus-like particle produced in situ during MV-infection. All recombinant MV revealed normal growths, genetic stability, and proper expression and presentation of both antigens. Potent antigen-specific humoral and cellular immunity were found in immunized MV-susceptible IFNAR-/--CD46Ge mice. These immune responses significantly inhibited metastasis formation or increased therapeutic efficacy compared to control MV in respective novel in vivo tumor models using syngeneic B16-hCD46/mCLDN6 murine melanoma cells. These data indicate the potential of MV to trigger selected tumor antigen-specific immune responses on top of direct tumor lysis for enhanced efficacy.


Assuntos
Antígenos de Neoplasias/genética , Vacinas Anticâncer/imunologia , Vírus do Sarampo/genética , Melanoma Experimental/terapia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Animais , Apresentação de Antígeno , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Autoanticorpos/sangue , Autoanticorpos/metabolismo , Vacinas Anticâncer/genética , Vacinas Anticâncer/uso terapêutico , Linhagem Celular Tumoral , Chlorocebus aethiops , Claudinas/genética , Claudinas/imunologia , Claudinas/metabolismo , Imunidade Celular , Imunidade Humoral , Interferon gama/metabolismo , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Melanoma Experimental/imunologia , Camundongos , Camundongos Transgênicos , Terapia Viral Oncolítica , Ovalbumina/genética , Ovalbumina/imunologia , Vacinas de Partículas Semelhantes a Vírus/genética , Vacinas de Partículas Semelhantes a Vírus/uso terapêutico , Células Vero
17.
Nat Commun ; 3: 796, 2012 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-22531181

RESUMO

The large virus family Paramyxoviridae includes some of the most significant human and livestock viruses, such as measles-, distemper-, mumps-, parainfluenza-, Newcastle disease-, respiratory syncytial virus and metapneumoviruses. Here we identify an estimated 66 new paramyxoviruses in a worldwide sample of 119 bat and rodent species (9,278 individuals). Major discoveries include evidence of an origin of Hendra- and Nipah virus in Africa, identification of a bat virus conspecific with the human mumps virus, detection of close relatives of respiratory syncytial virus, mouse pneumonia- and canine distemper virus in bats, as well as direct evidence of Sendai virus in rodents. Phylogenetic reconstruction of host associations suggests a predominance of host switches from bats to other mammals and birds. Hypothesis tests in a maximum likelihood framework permit the phylogenetic placement of bats as tentative hosts at ancestral nodes to both the major Paramyxoviridae subfamilies (Paramyxovirinae and Pneumovirinae). Future attempts to predict the emergence of novel paramyxoviruses in humans and livestock will have to rely fundamentally on these data.


Assuntos
Quirópteros/virologia , Reservatórios de Doenças/virologia , Mamíferos/virologia , Infecções por Paramyxoviridae/virologia , Paramyxoviridae/classificação , Paramyxoviridae/isolamento & purificação , Animais , Cães , Humanos , Camundongos , Dados de Sequência Molecular , Paramyxoviridae/genética , Filogenia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA