Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Antiviral Res ; 79(1): 49-61, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18329738

RESUMO

Cytomegalovirus infection is associated with severe disease in immunocompromised individuals. Current antiviral therapy faces several limitations. In a search of novel drug candidates, we describe here the anti-cytomegaloviral properties of two compounds of the chemical class of quinazolines, gefitinib (Iressa) and Ax7396 (RGB-315389). Both compounds showed strong inhibitory effects in vitro against human and animal cytomegaloviruses with IC(50)s in a low micromolar range. Cytotoxicity did not occur at these effective concentrations. The antiviral mode of action was based on the inhibition of protein kinase activity, mainly directed to a viral target kinase (UL97/M97) in addition to cellular target candidates. This was demonstrated by a high sensitivity of the respective protein kinases in vitro and by infection experiments with viral mutants carrying genomic alterations in the ORF UL97/M97 modulating viral drug sensitivity. In a guinea pig model, gefitinib showed inhibition of cytomegaloviral loads in blood and lung tissue. Importantly, the rate of mortality of infected animals was reduced by gefitinib treatment. In contrast to the in vitro data, Ax7396 showed no significant antiviral activity in a mouse model. Further in vivo analyses have to assess the potential use of gefitinib in the treatment of cytomegalovirus disease.


Assuntos
Antivirais/farmacologia , Citomegalovirus/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Animais , Antivirais/química , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citomegalovirus/enzimologia , Citomegalovirus/genética , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/tratamento farmacológico , Infecções por Citomegalovirus/fisiopatologia , Infecções por Citomegalovirus/virologia , Feminino , Cobaias , Humanos , Concentração Inibidora 50 , Masculino , Camundongos , Fosfotransferases/antagonistas & inibidores , Fosfotransferases/metabolismo , Inibidores de Proteínas Quinases/química , Quinazolinas/química , Ratos , Deleção de Sequência , Carga Viral , Ensaio de Placa Viral , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo
2.
Antiviral Res ; 69(2): 60-9, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16325931

RESUMO

Treatment of human cytomegalovirus (HCMV) infections with any of the currently available antiviral agents is frequently associated with the occurrence of severe complications, seriously threatening the successful outcome of treatment. Therefore, the development of novel antiviral strategies is a challenging goal of current investigations. Previously, we reported that artesunate (ART) is an effective, non-cytotoxic inhibitor of HCMV in vitro. Here, we demonstrate that the efficacy of the antiviral effect of ART is augmented by co-treatment of HCMV-infected fibroblasts with ferrous iron, i.e. Ferrosanol, and/or the iron transfer-mediating molecule holo-transferrin. This could alleviate the HCMV-induced modulation of cell surface expression of adhesion molecule Thy-1, suggesting that ART might be able to prevent pro-inflammatory effects of infection. The iron-enhanced, antiviral effect of ART could also be demonstrated in cultured cells infected with rat cytomegalovirus. Experiments using the RCMV/rat model showed that both the viral DNA load and virus titers in the salivary glands from infected rats were significantly reduced upon treatment with ART. Furthermore, an additive antiviral effect for ART together with each one of conventional anti-HCMV drugs, i.e. ganciclovir, cidofovir or foscarnet, was detected in HCMV-infected fibroblasts. These findings might open new perspectives regarding the use of ART in clinical trials.


Assuntos
Antimaláricos/uso terapêutico , Artemisininas/uso terapêutico , Infecções por Citomegalovirus/tratamento farmacológico , Citomegalovirus/efeitos dos fármacos , Infecções por Herpesviridae/tratamento farmacológico , Muromegalovirus/efeitos dos fármacos , Sesquiterpenos/uso terapêutico , Replicação Viral/efeitos dos fármacos , Animais , Antimaláricos/farmacologia , Antivirais/farmacologia , Antivirais/uso terapêutico , Artemisininas/farmacologia , Artesunato , Células Cultivadas , Infecções por Citomegalovirus/virologia , Quimioterapia Combinada , Compostos Ferrosos/uso terapêutico , Fibroblastos/virologia , Infecções por Herpesviridae/virologia , Humanos , Hospedeiro Imunocomprometido , Masculino , Muromegalovirus/fisiologia , Ratos , Ratos Endogâmicos Lew , Sesquiterpenos/farmacologia , Organismos Livres de Patógenos Específicos , Transferrina/uso terapêutico
3.
Transpl Immunol ; 23(1-2): 59-64, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20394817

RESUMO

BACKGROUND: Rodent models are a very helpful tool to investigate immunological mechanisms in allograft rejection. The aim of this study was to compare two different immunodeficient recipients in a humanized mouse model of arterial xenotransplantation in terms of reconstitution of the human immune system and rejection of the arterial graft. METHODS: Side branches of human mammary artery were transplanted as infrarenal aortic interposition grafts into C.B-17-SCID beige and C57BL/6-Rag2(-/-)gammac(-/-) recipients. 7days after surgery mice were reconstituted with 5x10(7) human peripheral blood mononuclear cells (hu PBMCs) and 30days after reconstitution mice were sacrificed and histologic analysis was performed. Peripheral blood and splenocytes were investigated by FACS and ELISA analysis to ensure engraftment of human CD45(+) cells. RESULTS: Transplant arteriosclerosis developed in non-PBMC-reconstituted C.B-17-SCID beige mice (intimal proliferation: 36.31+/-4.37%), but significantly less in C57BL/6-Rag2(-/-) gammac(-/-) recipients (intimal proliferation: 12.26+/-5.21%). After reconstitution with 5x10(7) unfractionated human PBMCs both mouse strains showed intima proliferation 30days after reconstitution (C.B-17-SCID beige: 28.49+/-7.95% and C57BL/6-Rag2(-/-) gammac(-/-): 44.58+/-11.08%). Whereas only very few human CD45(+) cells were found in mouse blood and spleen of C.B-17-SCID beige mice, C57BL/6-Rag2(-/-) gammac(-/-) mice revealed a reliable reconstitution. In addition, levels of human IgG and IgM within the peripheral blood were markedly higher in C57BL/6-Rag2(-/-) gammac(-/-) recipients. CONCLUSION: In this study we can show, that the use of C57BL/6-Rag2(-/-) gammac(-/-) mice may be advantageous compared to C.B-17-SCID beige recipients in a humanized mouse model of vessel transplantation.


Assuntos
Implante de Prótese Vascular/métodos , Proteínas de Ligação a DNA/genética , Rejeição de Enxerto/imunologia , Sistema Imunitário/imunologia , Leucócitos Mononucleares/imunologia , Transplante Homólogo/imunologia , Animais , Humanos , Antígenos Comuns de Leucócito/metabolismo , Leucócitos Mononucleares/citologia , Artéria Torácica Interna/transplante , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Modelos Animais , Transplante Homólogo/métodos
4.
J Virol ; 80(16): 8006-18, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16873257

RESUMO

Several viruses, including human cytomegalovirus (HCMV), encode proteins that colocalize with a cellular subnuclear structure known as ND10. Since only viral DNA deposited at ND10 initiates transcription, ND10 structures were hypothesized to be essential for viral replication. On the other hand, interferon treatment induces an up-regulation of ND10 structures and viruses have evolved polypeptides that disperse the dot-like accumulation of ND10 proteins, suggesting that ND10 could also be part of an intrinsic defense mechanism. In order to obtain evidence for either a proviral or an antiviral function of ND10, we generated primary human fibroblasts with a stable, short interfering RNA-mediated knockdown (kd) of PML. In these cells, other ND10-associated proteins like hDaxx showed a diffuse nuclear distribution. Interestingly, we observed that HCMV infection induced the de novo formation of ND10-like hDaxx and Sp100 accumulations that colocalized with IE2 and were disrupted, in the apparent absence of PML, in an IE1-dependent manner during the first hours after infection. Furthermore, infection of PML-kd cells with wild-type HCMV at a low multiplicity of infection resulted in enhanced replication. In particular, a significantly increased plaque formation was detected, suggesting that more cells are able to support initiation of replication in the absence of PML. While there was no difference in viral DNA uptake between PML-kd and control cells, we observed a considerable increase in the number of immediate-early (IE) protein-positive cells, indicating that the depletion of PML augments the initiation of viral IE gene expression. These results strongly suggest that PML functions as part of an intrinsic immune mechanism against cytomegalovirus infections.


Assuntos
Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/metabolismo , Citomegalovirus/fisiologia , Regulação Viral da Expressão Gênica , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Replicação Viral/genética , Proteínas Adaptadoras de Transdução de Sinal/análise , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antígenos Nucleares/análise , Antígenos Nucleares/metabolismo , Autoantígenos/análise , Autoantígenos/metabolismo , Núcleo Celular/química , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Proteínas Correpressoras , Fibroblastos/química , Fibroblastos/metabolismo , Fibroblastos/virologia , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Chaperonas Moleculares , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas Nucleares/análise , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Proteína da Leucemia Promielocítica , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo
5.
J Biol Chem ; 280(39): 33357-67, 2005 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-15975922

RESUMO

Replication of human cytomegalovirus is limited at the level of nucleocytoplasmic transport of viral capsids, a process that requires the disassembly of the nuclear lamina. Deletion of the protein kinase gene UL97 from the viral genome showed that the activity of pUL97 plays an important role for viral capsid egress. Here, we report that p32, a novel cellular interactor of the viral kinase pUL97, promotes the accumulation of pUL97 at the nuclear membrane by recruiting the p32-pUL97 complex to the lamin B receptor. Transfection of active pUL97, but not a catalytically inactive mutant, induced a redistribution of lamina components as demonstrated for recombinant lamin B receptor-green fluorescent protein and endogenous lamins A and C. Consistent with this, p32 itself and lamins were phosphorylated by pUL97. Importantly, overexpression of p32 in human cytomegalovirus-infected cells resulted in increased efficiency of viral replication and release of viral particles. Thus, it is highly suggestive that the cellular protein p32 recruits pUL97 to induce a dissolution of the nuclear lamina thereby facilitating the nuclear export of viral capsids.


Assuntos
Proteínas de Transporte/metabolismo , Citomegalovirus/enzimologia , Proteínas Mitocondriais/metabolismo , Lâmina Nuclear/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Células Cultivadas , Células Clonais , Citomegalovirus/metabolismo , Fibroblastos/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/análise , Pele/citologia , Técnicas do Sistema de Duplo-Híbrido , Replicação Viral
6.
J Gen Virol ; 85(Pt 7): 1995-2000, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15218185

RESUMO

The deregulation of cellular adhesion molecules by human cytomegalovirus (HCMV) appears to be correlated with the development of vascular disease. In this study, it was investigated whether the expression of Thy-1 (CD90), a member of the immunoglobulin superfamily of adhesion molecules with constitutive expression on fibroblast cells, is modulated following infection with HCMV. It was observed that Thy-1 cell surface expression decreased significantly during the course of infection. Addition of neutralizing antibodies, as well as UV inactivation of virus, prevented Thy-1 downregulation. In contrast, inhibition of virus replication by cidofovir did not alter Thy-1 regulation by HCMV, indicating that immediate-early (IE) and/or early (E) gene products are responsible. Interestingly, after infection of fibroblasts with a recombinant GFP-expressing virus, infected as well as non-infected cells showed a reduced Thy-1 cell surface expression. From these findings, it is concluded that IE or E gene products of HCMV induce a so far unidentified soluble factor that mediates Thy-1 downregulation.


Assuntos
Citomegalovirus/imunologia , Fibroblastos/imunologia , Regulação Viral da Expressão Gênica/imunologia , Antígenos Thy-1/genética , Antígenos CD/genética , Sequência de Bases , Primers do DNA , Amplificação de Genes/imunologia , Humanos , RNA Mensageiro/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA