Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Front Cell Dev Biol ; 10: 1063692, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36578787

RESUMO

The Raf/MEK/ERK signaling pathway plays a key role in regulating cellular proliferation, differentiation, apoptosis, cytokine production, and immune responses. However, it is also involved in diseases such as cancer, and numerous viruses rely on an active Raf/MEK/ERK pathway for propagation. This pathway, and particularly MEK1/2, are therefore promising therapeutic targets. Assessment of target engagement is crucial to determine pharmacodynamics or the efficacy of a MEK1/2 inhibitor. In the field of infectious diseases, this is usually first determined in clinical trials with healthy volunteers. One method to detect MEK1/2 inhibitor target engagement is to assess the degree of ERK1/2 phosphorylation, as ERK1/2 is the only known substrate of MEK1/2. As healthy subjects, however, only feature a low baseline MEK1/2 activation and therefore low ERK1/2 phosphorylation in most tissues, assessing target engagement is challenging, and robust methods are urgently needed. We hence developed a method using PBMCs isolated from whole blood of healthy blood donors, followed by ex vivo treatment with the MEK1/2 inhibitor zapnometinib and stimulation with PMA to first inhibit and then induce MEK1/2 activation. As PMA cannot activate MEK1/2 upon MEK1/2 inhibition, MEK1/2 inhibition results in impaired MEK1/2 activation. In contrast, PMA stimulation without MEK1/2 inhibition results in high MEK1/2 activation. We demonstrated that, without MEK1/2 inhibitor treatment, MEK1/2 stimulation with PMA induces high MEK1/2 activation, which is clearly distinguishable from baseline MEK1/2 activation in human PBMCs. Furthermore, we showed that treatment with the MEK1/2 inhibitor zapnometinib maintains the MEK1/2 activation at approximately baseline level despite subsequent stimulation with PMA. As our protocol is easy to follow and preserves the cells in an in vivo-like condition throughout the whole handling process, this approach can be a major advance for the easy assessment of MEK1/2 inhibitor target engagement in healthy probands for clinical drug development.

2.
Cell Mol Life Sci ; 79(1): 65, 2022 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013790

RESUMO

Coronavirus disease 2019 (COVID-19), the illness caused by a novel coronavirus now called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has led to more than 260 million confirmed infections and 5 million deaths to date. While vaccination is a powerful tool to control pandemic spread, medication to relieve COVID-19-associated symptoms and alleviate disease progression especially in high-risk patients is still lacking. In this study, we explore the suitability of the rapid accelerated fibrosarcoma/mitogen-activated protein kinase/extracellular signal-regulated kinase (Raf/MEK/ERK) pathway as a druggable target in the treatment of SARS-CoV-2 infections. We find that SARS-CoV-2 transiently activates Raf/MEK/ERK signaling in the very early infection phase and that ERK1/2 knockdown limits virus replication in cell culture models. We demonstrate that ATR-002, a specific inhibitor of the upstream MEK1/2 kinases which is currently evaluated in clinical trials as an anti-influenza drug, displays strong anti-SARS-CoV-2 activity in cell lines as well as in primary air-liquid-interphase epithelial cell (ALI) cultures, with a safe and selective treatment window. We also observe that ATR-002 treatment impairs the SARS-CoV-2-induced expression of pro-inflammatory cytokines, and thus might prevent COVID-19-associated hyperinflammation, a key player in COVID-19 progression. Thus, our data suggest that the Raf/MEK/ERK signaling cascade may represent a target for therapeutic intervention strategies against SARS-CoV-2 infections and that ATR-002 is a promising candidate for further drug evaluation.


Assuntos
Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , Fenamatos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , SARS-CoV-2/efeitos dos fármacos , Células A549 , Adulto , Animais , COVID-19/metabolismo , Linhagem Celular , Células Cultivadas , Chlorocebus aethiops , Citocinas/metabolismo , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/antagonistas & inibidores , MAP Quinase Quinase 2/metabolismo , SARS-CoV-2/fisiologia , Células Vero , Replicação Viral/efeitos dos fármacos
3.
J Proteome Res ; 20(1): 289-304, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33141586

RESUMO

To understand and treat immunology-related diseases, a comprehensive, unbiased characterization of major histocompatibility complex (MHC) peptide ligands is of key importance. Preceding the analysis by mass spectrometry, MHC class I peptide ligands are typically isolated by MHC immunoaffinity chromatography (MHC-IAC) and less often by mild acid elution (MAE). MAE may provide a cheap alternative to MHC-IAC for suspension cells but has been hampered by the high number of contaminating, MHC-unrelated peptides. Here, we optimized MAE, yielding MHC peptide ligand purities of more than 80%. When compared with MHC-IAC, obtained peptides were similar in numbers, identities, and to a large extent intensities, while the percentage of cysteinylated peptides was 5 times higher in MAE. The latter benefitted the discovery of MHC-allotype-specific, distinct cysteinylation frequencies at individual positions of MHC peptide ligands. MAE revealed many MHC ligands with unmodified, N-terminal cysteine residues which get lost in MHC-IAC workflows. The results support the idea that MAE might be particularly valuable for the high-confidence analysis of post-translational modifications by avoiding the exposure of the investigated peptides to enzymes and reactive molecules in the cell lysate. Our improved and carefully documented MAE workflow represents a high-quality, cost-effective alternative to MHC-IAC for suspension cells.


Assuntos
Cisteína , Peptídeos , Cromatografia de Afinidade , Antígenos de Histocompatibilidade Classe I/metabolismo , Espectrometria de Massas , Ligação Proteica
4.
EMBO Mol Med ; 12(5): e10938, 2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-32163240

RESUMO

The current seasonal inactivated influenza vaccine protects only against a narrow range of virus strains as it triggers a dominant antibody response toward the hypervariable hemagglutinin (HA) head region. The discovery of rare broadly protective antibodies against conserved regions in influenza virus proteins has propelled research on distinct antigens and delivery methods to efficiently induce broad immunity toward drifted or shifted virus strains. Here, we report that adeno-associated virus (AAV) vectors expressing influenza virus HA or chimeric HA protected mice against homologous and heterologous virus challenges. Unexpectedly, immunization even with wild-type HA induced antibodies recognizing the HA-stalk and activating FcγR-dependent responses indicating that AAV-vectored expression balances HA head- and HA stalk-specific humoral responses. Immunization with AAV-HA partially protected also ferrets against a harsh virus challenge. Results from this study provide a rationale for further clinical development of AAV vectors as influenza vaccine platform, which could benefit from their approved use in human gene therapy.


Assuntos
Vacinas contra Influenza , Influenza Humana , Infecções por Orthomyxoviridae , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , Dependovirus/genética , Furões , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Humanos , Influenza Humana/prevenção & controle , Camundongos , Infecções por Orthomyxoviridae/prevenção & controle
5.
Sci Data ; 5: 180157, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30084848

RESUMO

The large array of peptides presented to CD8+ T cells by major histocompatibility complex (MHC) class I molecules is referred to as the MHC class I immunopeptidome. Although the MHC class I immunopeptidome is ubiquitous in mammals and represents a critical component of the immune system, very little is known, in any species, about its composition across most tissues and organs in vivo. We applied mass spectrometry (MS) technologies to draft the first tissue-based atlas of the murine MHC class I immunopeptidome in health. Peptides were extracted from 19 normal tissues from C57BL/6 mice and prepared for MS injections, resulting in a total number of 28,448 high-confidence H2Db/Kb-associated peptides identified and annotated in the atlas. This atlas provides initial qualitative data to explore the tissue-specificity of the immunopeptidome and serves as a guide to identify potential tumor-associated antigens from various cancer models. Our data were shared via PRIDE (PXD008733), SysteMHC Atlas (SYSMHC00018) and SWATH Atlas. We anticipate that this unique dataset will be expanded in the future and will find wide applications in basic and translational immunology.


Assuntos
Antígenos de Histocompatibilidade Classe I , Especificidade de Órgãos , Animais , Antígenos de Histocompatibilidade Classe I/análise , Antígenos de Histocompatibilidade Classe I/imunologia , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos
6.
Antiviral Res ; 157: 80-92, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29990517

RESUMO

Influenza A virus (IAV) infections are still a major global threat for humans, especially for the risk groups of young children and the elderly. Annual epidemics and sporadically occurring pandemics highlight the necessity of effective antivirals that can limit viral replication. The currently licensed antiviral drugs target viral factors and are prone to provoke viral resistance. In infected host cells IAV induces various cellular signaling cascades. The Raf/MEK/ERK signaling cascade is indispensable for IAV replication because it triggers the nuclear export of newly assembled viral ribonucleoproteins (vRNPs). Inhibition of this cascade limits viral replication. Thus, next to their potential in anti-tumor therapy, inhibitors targeting the Raf/MEK/ERK signaling cascade came into focus as potential antiviral drugs. The first licensed MEK inhibitor Trametinib (GSK-1120212) is used for treatment of malignant melanoma, being highly selective and having a promising side effect profile. Since Trametinib may be qualified for a repurposing approach that would significantly shorten development time for an anti-flu use, we evaluated its antiviral potency and mode of action. In this study, we describe that Trametinib efficiently blocks replication of different IAV subtypes in vitro and in vivo. The broad antiviral activity against various IAV strains was due to its ability to interfere with export of progeny vRNPs from the nucleus. The compound also limited hyper-expression of several cytokines. Thus, we show for the first time that a clinically approved MEK inhibitor acts as a potent anti-influenza agent.


Assuntos
Antivirais/farmacologia , Citocinas/antagonistas & inibidores , Fatores Imunológicos/farmacologia , Vírus da Influenza A/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Piridonas/farmacologia , Pirimidinonas/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Linhagem Celular , Reposicionamento de Medicamentos , Humanos , Vírus da Influenza A/crescimento & desenvolvimento
7.
Infect Immun ; 85(12)2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28893917

RESUMO

Influenza A virus (IAV) infection is often followed by secondary bacterial lung infection, which is a major reason for severe, often fatal pneumonia. Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) strains such as USA300 cause particularly severe and difficult-to-treat cases of IAV-associated pneumonia. CA-MRSA strains are known to produce extraordinarily large amounts of phenol-soluble modulin (PSM) peptides, which are important cytotoxins and proinflammatory molecules that contribute to several types of S. aureus infection. However, their potential role in pneumonia has remained elusive. We determined the impact of PSMs on human lung epithelial cells and found that PSMs are cytotoxic and induce the secretion of the proinflammatory cytokine interleukin-8 (IL-8) in these cells. Both effects were boosted by previous infection with the 2009 swine flu pandemic IAV H1N1 strain, suggesting that PSMs may contribute to lung inflammation and damage in IAV-associated S. aureus pneumonia. Notably, the PSM-producing USA300 strain caused a higher mortality rate than did an isogenic PSM-deficient mutant in a mouse IAV-S. aureus pneumonia coinfection model, indicating that PSMs are major virulence factors in IAV-associated S. aureus pneumonia and may represent important targets for future anti-infective therapies.


Assuntos
Toxinas Bacterianas/metabolismo , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Influenza Humana/complicações , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Pneumonia Estafilocócica/patologia , Fatores de Virulência/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Células Epiteliais/microbiologia , Células Epiteliais/fisiologia , Humanos , Influenza Humana/virologia , Staphylococcus aureus Resistente à Meticilina/metabolismo , Camundongos , Análise de Sobrevida , Suínos
8.
PLoS One ; 11(11): e0167017, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27893789

RESUMO

There are limitations in pre-clinical settings using mice as a basis for clinical development in humans. In cancer, similarities exist between humans and dogs; thus, the dog patient can be a link in the transition from laboratory research on mouse models to clinical trials in humans. Knowledge of the peptides presented on MHC molecules is fundamental for the development of highly specific T cell-based immunotherapies. This information is available for human MHC molecules but is absent for the canine MHC. In the present study, we characterized the binding motif of dog leukocyte antigen (DLA) class I allele DLA-88*50101, using human C1R and K562 transfected cells expressing the DLA-88*50101 heavy chain. MHC class I immunoaffinity-purification revealed 3720 DLA-88*50101 derived peptides, which enabled the determination of major anchor positions. The characterized binding motif of DLA-88*50101 was similar to HLA-A*02:01. Peptide binding analyses on HLA-A*02:01 and DLA-88*50101 via flow cytometry showed weak binding of DLA-88*50101 derived peptides to HLA-A*02:01, and vice versa. Our results present for the first time a detailed peptide binding motif of the canine MHC class I allelic product DLA-88*50101. These data support the goal of establishing dogs as a suitable animal model for the evaluation and development of T cell-based cancer immunotherapies, benefiting both dog and human patients.


Assuntos
Doenças do Cão/imunologia , Doenças do Cão/terapia , Antígenos de Histocompatibilidade Classe I/imunologia , Imunoterapia , Fragmentos de Peptídeos/imunologia , Linfócitos T/imunologia , Sequência de Aminoácidos , Animais , Cães , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Células K562 , Camundongos , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Polimorfismo Genético/genética , Ligação Proteica , Homologia de Sequência de Aminoácidos
9.
PLoS One ; 8(5): e63657, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23717460

RESUMO

Infections with influenza A viruses (IAV) are still amongst the major causes of highly contagious severe respiratory diseases not only bearing a devastating effect to human health, but also significantly impact the economy. Besides vaccination that represents the best option to protect from IAV infections, only two classes of anti-influenza drugs, inhibitors of the M2 ion channel and the neuraminidase, often causing resistant IAV variants have been approved. That is why the need for effective and amply available antivirals against IAV is of high priority. Here we introduce LADANIA067 from the leaves of the wild black currant (Ribes nigrum folium) as a potent compound against IAV infections in vitro and in vivo. LADANIA067 treatment resulted in a reduction of progeny virus titers in cell cultures infected with prototype avian and human influenza virus strains of different subtypes. At the effective dose of 100 µg/ml the extract did not exhibit apparent harming effects on cell viability, metabolism or proliferation. Further, viruses showed no tendency to develop resistance to LADANIA067 when compared to amantadine that resulted in the generation of resistant variants after only a few passages. On a molecular basis the protective effect of LADANIA067 appears to be mainly due to interference with virus internalisation. In the mouse infection model LADANIA067 treatment reduces progeny virus titers in the lung upon intranasal application. In conclusion, an extract from the leaves of the wild black currant might be a promising source for the development of new antiviral compounds to fight IAV infections.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Extratos Vegetais/farmacologia , Folhas de Planta/química , Ribes/química , Internalização do Vírus/efeitos dos fármacos , Animais , Antivirais/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células , Cães , Avaliação Pré-Clínica de Medicamentos , Farmacorresistência Viral/efeitos dos fármacos , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Vírus da Influenza A Subtipo H1N1/fisiologia , Vírus da Influenza A Subtipo H7N7/efeitos dos fármacos , Influenza Humana/tratamento farmacológico , Influenza Humana/virologia , Camundongos , Camundongos Endogâmicos BALB C , Extratos Vegetais/uso terapêutico , Replicação Viral/efeitos dos fármacos
10.
Antiviral Res ; 98(2): 319-24, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23523553

RESUMO

MEK inhibitors are very potent and promising compounds in cancer therapy. Earlier investigations have demonstrated that they also possess antiviral properties against influenza virus. This is due to the fact that activation of the Raf/MEK/ERK signaling pathway is a prerequisite for influenza virus replication. As an alternative to vaccination, antiviral therapy is a means to control influenza. The appearance of influenza virus strains that are resistant to current treatment options demonstrates the need for new antiviral strategies. The aim of the presented study was to investigate whether the combination of MEK inhibitors with oseltamivir, an inhibitor of viral neuraminidase activity, would result in a synergistic antiviral effect against pandemic influenza A/Regensburg/D6/2009 (H1N1pdm09) virus. Here we show that four different MEK inhibitors, PD-0325901, AZD-6244, AZD-8330 and RDEA-119 that are orally available and at least in a phase I clinical trial against cancer demonstrate antiviral activity as single agents or in combination with oseltamivir. Combination treatment increased the antiviral activity of oseltamivir significantly and resulted in a synergistic antiviral effect as determined by the Chou-Talalay method. Taken together, the results demonstrate increased antiviral activity of oseltamivir after combination with MEK inhibitors. These data are promising for further preclinical in vitro and in vivo investigations on the way to developing new antiviral regimens against influenza.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Influenza Humana/virologia , MAP Quinase Quinase 1/antagonistas & inibidores , Oseltamivir/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Vírus da Influenza A Subtipo H1N1/fisiologia , Influenza Humana/tratamento farmacológico
11.
J Virol ; 86(18): 10211-7, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22787206

RESUMO

Influenza A virus (IAV) infection of epithelial cells activates NF-κB transcription factors via the canonical NF-κB signaling pathway, which modulates both the antiviral immune response and viral replication. Since almost nothing is known so far about a function of noncanonical NF-κB signaling after IAV infection, we tested infected cells for activation of p52 and RelB. We show that the viral NS1 protein strongly inhibits RIG-I-mediated noncanonical NF-κB activation and expression of the noncanonical target gene CCL19.


Assuntos
RNA Helicases DEAD-box/antagonistas & inibidores , Vírus da Influenza A/fisiologia , NF-kappa B/metabolismo , Proteínas não Estruturais Virais/fisiologia , Linhagem Celular , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Vírus da Influenza A/patogenicidade , Pulmão/metabolismo , Pulmão/virologia , Subunidade p52 de NF-kappa B/metabolismo , Receptores Imunológicos , Transdução de Sinais , Fator de Transcrição RelB/metabolismo
12.
Cell Microbiol ; 14(7): 1135-47, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22417706

RESUMO

The innate immune response of influenza A virus-infected cells is predominantly mediated by type I interferon-induced proteins. Expression of the interferon ß (IFNß) itself is initiated by accumulating viral RNA and is transmitted by different signalling cascades that feed into activation of the three transcriptional elements located in the IFNß promoter, AP-1, IRF-3 and NF-κB. FHL2 (four-and-a-half LIM domain protein 2) is an adaptor molecule that shuttles between membrane and nucleus regulating signalling cascades and gene transcription. Here we describe FHL2 as a novel regulator of influenza A virus propagation. Using mouse FHL2 wild-type, knockout and rescued cells and human epithelial cells with different expression levels of FHL2 we showed that FHL2 decreases influenza A virus propagation by regulating the intrinsic cellular antiviral immune response. On virus infection FHL2 translocates into the nucleus, potentiating the IRF-3-dependent transcription of the IFNß gene.


Assuntos
Imunidade Inata , Vírus da Influenza A/imunologia , Influenza Humana/imunologia , Proteínas com Homeodomínio LIM/metabolismo , Proteínas Musculares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Células Epiteliais/virologia , Regulação da Expressão Gênica , Humanos , Fator Regulador 3 de Interferon/biossíntese , Camundongos , Camundongos Knockout
13.
Antiviral Res ; 91(3): 304-13, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21777621

RESUMO

The appearance of highly pathogenic avian influenza A viruses of the H5N1 subtype being able to infect humans and the 2009 H1N1 pandemic reveals the urgent need for new and efficient countermeasures against these viruses. The long-term efficacy of current antivirals is often limited, because of the emergence of drug-resistant virus mutants. A growing understanding of the virus-host interaction raises the possibility to explore alternative targets involved in the viral replication. In the present study we show that the proteasome inhibitor VL-01 leads to reduction of influenza virus replication in human lung adenocarcinoma epithelial cells (A549) as demonstrated with three different influenza virus strains, A/Puerto Rico/8/34 (H1N1) (EC50 value of 1.7 µM), A/Regensburg/D6/09 (H1N1v) (EC50 value of 2.4 µM) and A/Mallard/Bavaria/1/2006 (H5N1) (EC50 value of 0.8 µM). In in vivo experiments we could demonstrate that VL-01-aerosol-treatment of BALB/c mice with 14.1 mg/kg results in no toxic side effects, reduced progeny virus titers in the lung (1.1 ± 0.3 log10 pfu) and enhanced survival of mice after infection with a 5-fold MLD50 of the human influenza A virus strain A/Puerto Rico/8/34 (H1N1) up to 50%. Furthermore, treatment of mice with VL-01 reduced the cytokine release of IL-α/ß, IL-6, MIP-1ß, RANTES and TNF-α induced by LPS or highly pathogen avian H5N1 influenza A virus. The present data demonstrates an antiviral effect of VL-01 in vitro and in vivo and the ability to reduce influenza virus induced cytokines and chemokines.


Assuntos
Citocinas/antagonistas & inibidores , Inibidores Enzimáticos/administração & dosagem , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Infecções por Orthomyxoviridae/tratamento farmacológico , Inibidores de Proteassoma , Replicação Viral/efeitos dos fármacos , Administração por Inalação , Animais , Antivirais/administração & dosagem , Antivirais/uso terapêutico , Linhagem Celular Tumoral , Citocinas/biossíntese , Farmacorresistência Viral , Inibidores Enzimáticos/uso terapêutico , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Virus da Influenza A Subtipo H5N1/crescimento & desenvolvimento , Influenza Humana/tratamento farmacológico , Influenza Humana/virologia , Lipopolissacarídeos/efeitos adversos , Lipopolissacarídeos/farmacologia , Pulmão/citologia , Pulmão/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/virologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Taxa de Sobrevida
14.
Antiviral Res ; 92(1): 45-56, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21641936

RESUMO

Influenza virus (IV) infection can cause severe pneumonia and death. Therapeutic actions are limited to vaccines and a few anti-viral drugs. These target viral functions thereby selecting resistant variants. During replication IV activates the Raf/MEK/ERK-cascade and the transcription factor NF-kappaB. Both result in virus supportive and anti-viral effects by promoting viral genome transport for virus assembly and by inducing expression of pro-inflammatory host factors. Apart from tissue damage caused by the virus lytic replication, an imbalanced overproduction of anti-viral cytokines can cause severe lung damage as observed in human H5-type IV infections. Recently we showed that inhibition of NF-kappaB activity reduces the virus titer in vitro and in vivo. We have now analyzed whether inhibition of these pathways, allows simultaneous reduction of virus titers and virus-induced cytokines. The results show that inhibition of either pathway indeed leads to decreased virus titers and cytokine expression. This was not only true for infected permanent cells or primary mouse alveolar epithelial cells, but also in infected mice. Hereby we demonstrate for the first time in vitro and in vivo that virus titers and pro-inflammatory cytokine expression can be modulated simultaneously. This could provide a new rationale of future therapeutic strategies to treat IV pneumonia.


Assuntos
Citocinas/genética , Regulação para Baixo , Influenza Humana/metabolismo , Sistema de Sinalização das MAP Quinases , NF-kappa B/metabolismo , Orthomyxoviridae/fisiologia , Carga Viral , Animais , Antivirais/farmacologia , Linhagem Celular , Citocinas/biossíntese , Regulação para Baixo/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Influenza Humana/enzimologia , Influenza Humana/genética , Influenza Humana/virologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , Nitrilas/farmacologia , Orthomyxoviridae/efeitos dos fármacos , Orthomyxoviridae/genética , Proteínas Proto-Oncogênicas c-raf/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo , Sulfonas/farmacologia , Carga Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
15.
J Interferon Cytokine Res ; 31(6): 515-25, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21323570

RESUMO

The recent emergence of pandemic swine-origin influenza virus (H1N1) and the severe outbreaks of highly pathogenic avian influenza virus of the H5N1 subtype leading to death in humans is a reminder that influenza remains a frightening foe throughout the world. Besides vaccination, there is an urgent need for new antiviral strategies to protect against influenza. The innate immune response to influenza viruses involves production of interferon alpha and beta (IFN-α/ß), which plays a crucial role in virus clearance during the initial stage of infection. We examined the effect of IFN-α on the replication of H5N1 and H1N1 in vitro and in vivo. A single pretreatment with low-dose IFN-α reduced lung virus titers up to 1.4 log(10) pfu. The antiviral effect increased after multiple pretreatments. Low-dose IFN-α protected mice against lethal H5N1 viral infection. Further, IFN-α was also effective against H1N1 in vitro and in the mouse model. These results indicate that low-dose IFN-α treatment leads to the induction of antiviral cytokines that are involved in the reduction of influenza virus titers in the lung. Moreover, it might be possible that a medical application during pandemic outbreak could help contain fulminant infections.


Assuntos
Células Epiteliais/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/fisiologia , Virus da Influenza A Subtipo H5N1/fisiologia , Influenza Humana/tratamento farmacológico , Interferon Tipo I/administração & dosagem , Infecções por Orthomyxoviridae/tratamento farmacológico , Animais , Aves , Linhagem Celular Tumoral , Progressão da Doença , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Células Epiteliais/virologia , Fibroblastos/imunologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibroblastos/virologia , Humanos , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/patogenicidade , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Virus da Influenza A Subtipo H5N1/patogenicidade , Influenza Aviária/virologia , Influenza Humana/imunologia , Influenza Humana/virologia , Interferon Tipo I/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/fisiopatologia , Infecções por Orthomyxoviridae/virologia , Proteínas Recombinantes , Especificidade da Espécie , Suínos , Replicação Viral/efeitos dos fármacos
16.
J Pept Sci ; 17(3): 226-32, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21308879

RESUMO

The influenza virus, major surface glycoprotein hemagglutinin (HA) is one of the principal targets for the development of protective immunity. Aiming at contributing to the development of a vaccine that remains the first choice for prophylactic intervention, a reconstituted model of HA, mimicking its antigenic properties was designed, synthesized and tested in mice for the induction of protective immunity. Four helper T lymphocyte [HTL (T(1) , T(3) , T(7) and T(8) )] and four cytotoxic lymphocyte [CTL (T(2) , T(4) , T(5) and T(6) )] epitopes were coupled in two copies each to an artificial carrier, SOC(4) , which was formed by the repeating tripeptide Lys-Aib-Gly. The helical conformation of the SOC(4) -conjugates preserves the initial topology of the attached epitopes, which is critical for their immunogenic properties. Survival of immunized animals, ranged from 30 to 50%, points out the induction of protective immunity by using the SOC(4) -conjugates.


Assuntos
Epitopos de Linfócito T/imunologia , Hemaglutininas/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Peptídeos/síntese química , Peptídeos/imunologia , Animais , Cromatografia Líquida de Alta Pressão , Dicroísmo Circular , Epitopos de Linfócito T/química , Feminino , Espectrometria de Massas , Camundongos , Peptídeos/química
17.
Vaccine ; 29(8): 1690-9, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21195079

RESUMO

The recent H1N1 influenza pandemic and the inevitable delay between identification of the virus and production of the specific vaccine have highlighted the urgent need for new generation influenza vaccines that can preemptively induce broad immunity to different strains of the virus. In this study we have produced AAV-based vectors expressing the A/Mexico/4603/2009 (H1N1) hemagglutinin (HA), nucleocapsid (NP) and the matrix protein M1 and have evaluated their ability to induce specific immune response and protect mice against homologous and heterologous challenge. Each of the vaccine vectors elicited potent cellular and humoral immune responses in mice. Although immunization with AAV-M1 did not improve survival after challenge with the homologous strain, immunization with the AAV-H1 and AAV-NP vectors resulted in survival of all mice, as did inoculation with a combination of all three vectors. Furthermore, trivalent vaccination also conferred partial protection against challenge with the highly heterologous and virulent A/PR/8/34 strain of H1N1 influenza.


Assuntos
Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Feminino , Células HEK293 , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Imunidade Celular , Imunidade Humoral , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Testes de Neutralização , Proteínas do Nucleocapsídeo , Infecções por Orthomyxoviridae/imunologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/imunologia , Transfecção , Proteínas do Core Viral/genética , Proteínas do Core Viral/imunologia , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/imunologia , Ensaio de Placa Viral
18.
Am J Pathol ; 175(3): 1178-86, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19700749

RESUMO

SC35M is a mouse-adapted variant of the highly pathogenic avian influenza virus SC35. We have previously shown that interspecies adaptation is mediated by mutations in the viral polymerase and that it is paralleled by the acquisition of high pathogenicity for mice. In the present study, we have compared virus spread and organ tropism of SC35 and SC35M in mice. We show that SC35 virus causes mild bronchiolitis in these animals, whereas infection with the mouse-adapted SC35M virus leads to severe hemorrhagic pneumonia with dissemination to other organs, including the brain. In SC35M-infected animals, viral RNA and viral antigen were detected in monocytes and macrophages, and SC35M, unlike SC35, replicated in lymphocyte and macrophage cultures in vitro. SC35M did not induce an adequate cytokine response but, unlike SC35, caused severe lymphopenia in mice. These observations suggest that the high efficiency of the SC35M polymerase is responsible for infection and depletion of lymphocytes and other white blood cells, which results in immune suppression and systemic virus spread.


Assuntos
Terapia de Imunossupressão , Vírus da Influenza A/patogenicidade , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/fisiopatologia , Infecções por Orthomyxoviridae/virologia , Animais , Antígenos Virais/análise , Bronquiolite/etiologia , Bronquiolite/fisiopatologia , Células Cultivadas , Embrião de Galinha , Vírus da Influenza A Subtipo H7N7/genética , Vírus da Influenza A Subtipo H7N7/patogenicidade , Vírus da Influenza A/genética , Macrófagos/imunologia , Macrófagos/virologia , Camundongos , Monócitos/imunologia , Monócitos/virologia , Infecções por Orthomyxoviridae/complicações , Pneumonia Viral/etiologia , Pneumonia Viral/fisiopatologia , RNA Viral/análise
19.
Antiviral Res ; 76(1): 38-47, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17572513

RESUMO

Infections with influenza A viruses still pose a major threat to humans and several animal species. The occurrence of highly pathogenic avian influenza viruses of the H5N1 subtype capable to infect and kill humans highlights the urgent need for new and efficient countermeasures against this viral disease. Here we demonstrate that a polyphenol rich extract (CYSTUS052) from the Mediterranean plant Cistus incanus exerts a potent anti-influenza virus activity in A549 or MDCK cell cultures infected with prototype avian and human influenza strains of different subtypes. CYSTUS052 treatment resulted in a reduction of progeny virus titers of up to two logs. At the effective dose of 50 microg/ml the extract did not exhibit apparent harming effects on cell viability, metabolism or proliferation, which is consistent with the fact that these plant extracts are already used in traditional medicine in southern Europe for centuries without any reported complications. Viruses did not develop resistance to CYSTUS052 when compared to amantadine that resulted in the generation of resistant variants after only a few passages. On a molecular basis the protective effect of CYSTUS052 appears to be mainly due to binding of the polymeric polyphenol components of the extract to the virus surface, thereby inhibiting binding of the hemagglutinin to cellular receptors. Thus, a local application of CYSTUS052 at the viral entry routes may be a promising approach that may help to protect from influenza virus infections.


Assuntos
Antivirais/farmacologia , Citrus , Flavonoides , Vírus da Influenza A/efeitos dos fármacos , Influenza Aviária/virologia , Influenza Humana/virologia , Fenóis , Extratos Vegetais/farmacologia , Animais , Antivirais/química , Antivirais/toxicidade , Aves , Linhagem Celular , Cães , Farmacorresistência Viral , Humanos , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Vírus da Influenza A Subtipo H7N7/efeitos dos fármacos , Extratos Vegetais/química , Extratos Vegetais/toxicidade , Polifenóis , Inoculações Seriadas
20.
Antiviral Res ; 76(1): 1-10, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17573133

RESUMO

Influenza, a respiratory disease caused by influenza viruses, is still a worldwide threat with a high potential to cause a pandemic. Beside vaccination, only two classes of drugs are available for antiviral treatment against the pathogen. Here we show that CYSTUS052, a plant extract from a special variety of Cistus incanus that is rich in polymeric polyphenols, exhibits antiviral activity against a highly pathogenic avian influenza A virus (H7N7) in cell culture and in a mouse infection model. In vitro and in vivo treatment was performed with an aerosol formulation, because the bioavailability of high molecular weight polyphenols is poor. In MDCK cells, a 90% reduction of plaque numbers on cells pre-incubated with the plant extract was achieved. For in vivo experiments we used a novel monitoring system for influenza A virus-infected mice that allows measurement of body temperature and gross motor-activity of the animals. Mice treated with CYSTUS052 did not develop disease, showed neither differences in their body temperature nor differences in their gross motor-activity and exhibited no histological alterations of the bronchiolus epithelial cells.


Assuntos
Antivirais/química , Citrus , Flavonoides , Vírus da Influenza A Subtipo H7N7 , Infecções por Orthomyxoviridae/tratamento farmacológico , Fenóis , Extratos Vegetais/química , Aerossóis , Animais , Antivirais/administração & dosagem , Brônquios/patologia , Cães , Esquema de Medicação , Avaliação Pré-Clínica de Medicamentos , Células Epiteliais/patologia , Feminino , Febre/patologia , Vírus da Influenza A Subtipo H7N7/crescimento & desenvolvimento , Inalação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Movimento , Infecções por Orthomyxoviridae/fisiopatologia , Fenóis/administração & dosagem , Fenóis/química , Extratos Vegetais/administração & dosagem , Polifenóis , Ensaio de Placa Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA