Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
EMBO Mol Med ; 8(8): 831-8, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27311961

RESUMO

The mouse is the main model organism used to study the functions of human genes because most biological processes in the mouse are highly conserved in humans. Recent reports that compared identical transcriptomic datasets of human inflammatory diseases with datasets from mouse models using traditional gene-to-gene comparison techniques resulted in contradictory conclusions regarding the relevance of animal models for translational research. To reduce susceptibility to biased interpretation, all genes of interest for the biological question under investigation should be considered. Thus, standardized approaches for systematic data analysis are needed. We analyzed the same datasets using gene set enrichment analysis focusing on pathways assigned to inflammatory processes in either humans or mice. The analyses revealed a moderate overlap between all human and mouse datasets, with average positive and negative predictive values of 48 and 57% significant correlations. Subgroups of the septic mouse models (i.e., Staphylococcus aureus injection) correlated very well with most human studies. These findings support the applicability of targeted strategies to identify the optimal animal model and protocol to improve the success of translational research.


Assuntos
Modelos Animais de Doenças , Redes Reguladoras de Genes , Biologia de Sistemas/métodos , Pesquisa Translacional Biomédica/métodos , Animais , Perfilação da Expressão Gênica , Humanos , Camundongos , Sepse/genética , Sepse/patologia
2.
Circulation ; 130(18): 1589-600, 2014 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-25165091

RESUMO

BACKGROUND: Common causative agents in the development of inflammatory cardiomyopathy include cardiotropic viruses such as coxsackievirus B3 (CVB3). Here, we investigated the role of the ubiquitin-like modifier interferon-stimulated gene of 15 kDa (ISG15) in the pathogenesis of viral cardiomyopathy. METHODS AND RESULTS: In CVB3-infected mice, the absence of protein modification with ISG15 was accompanied by a profound exacerbation of myocarditis and by a significant increase in mortality and heart failure. We found that ISG15 in cardiomyocytes contributed significantly to the suppression of viral replication. In the absence of an intact ISG15 system, virus titers were markedly elevated by postinfection day 8, and viral RNA persisted in ISG15(-/-) mice at postinfection day 28. Ablation of the ISG15 protein modification system in CVB3 infection predisposed mice to long-term disease with deposition of collagen fibers, all leading to inflammatory cardiomyopathy. We found that ISG15 acts as part of the intrinsic immunity in cardiomyocytes and detected no significant effects of ISG15 modification on the cellular immune response. ISG15 modification of CVB3 2A protease counterbalanced CVB3-induced cleavage of the host cell eukaryotic initiation factor of translation eIF4G in cardiomyocytes, thereby counterbalancing the shutoff of host cell translation in CVB3 infection. We demonstrate that ISG15 suppressed infectious virus yield in human cardiac myocytes and the induction of ISG15 in patients with viral cardiomyopathy. CONCLUSIONS: The ISG15 conjugation system represents a critical innate response mechanism in cardiomyocytes to fight the battle against invading pathogens, limiting inflammatory cardiomyopathy, heart failure, and death. Interference with the ISG15 system might be a novel therapeutic approach in viral cardiomyopathy.


Assuntos
Cardiomiopatia Dilatada/virologia , Infecções por Coxsackievirus/complicações , Citocinas/genética , Enterovirus Humano B/imunologia , Insuficiência Cardíaca/virologia , Adulto , Animais , Biópsia , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/imunologia , Infecções por Coxsackievirus/genética , Infecções por Coxsackievirus/imunologia , Cisteína Endopeptidases/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/imunologia , Humanos , Imunidade Inata/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/virologia , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Miócitos Cardíacos/fisiologia , Miócitos Cardíacos/virologia , Linfócitos T/imunologia , Linfócitos T/virologia , Ubiquitinas/genética , Ubiquitinas/imunologia , Ubiquitinas/metabolismo , Proteínas Virais/imunologia , Replicação Viral
3.
PLoS Pathog ; 7(9): e1002233, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21909276

RESUMO

Proteasomes recognize and degrade poly-ubiquitinylated proteins. In infectious disease, cells activated by interferons (IFNs) express three unique catalytic subunits ß1i/LMP2, ß2i/MECL-1 and ß5i/LMP7 forming an alternative proteasome isoform, the immunoproteasome (IP). The in vivo function of IPs in pathogen-induced inflammation is still a matter of controversy. IPs were mainly associated with MHC class I antigen processing. However, recent findings pointed to a more general function of IPs in response to cytokine stress. Here, we report on the role of IPs in acute coxsackievirus B3 (CVB3) myocarditis reflecting one of the most common viral disease entities among young people. Despite identical viral load in both control and IP-deficient mice, IP-deficiency was associated with severe acute heart muscle injury reflected by large foci of inflammatory lesions and severe myocardial tissue damage. Exacerbation of acute heart muscle injury in this host was ascribed to disequilibrium in protein homeostasis in viral heart disease as indicated by the detection of increased proteotoxic stress in cytokine-challenged cardiomyocytes and inflammatory cells from IP-deficient mice. In fact, due to IP-dependent removal of poly-ubiquitinylated protein aggregates in the injured myocardium IPs protected CVB3-challenged mice from oxidant-protein damage. Impaired NFκB activation in IP-deficient cardiomyocytes and inflammatory cells and proteotoxic stress in combination with severe inflammation in CVB3-challenged hearts from IP-deficient mice potentiated apoptotic cell death in this host, thus exacerbating acute tissue damage. Adoptive T cell transfer studies in IP-deficient mice are in agreement with data pointing towards an effective CD8 T cell immune. This study therefore demonstrates that IP formation primarily protects the target organ of CVB3 infection from excessive inflammatory tissue damage in a virus-induced proinflammatory cytokine milieu.


Assuntos
Infecções por Coxsackievirus/imunologia , Enterovirus Humano B , Miocardite/imunologia , Complexo de Endopeptidases do Proteassoma/deficiência , Complexo de Endopeptidases do Proteassoma/imunologia , Subunidades Proteicas/deficiência , Animais , Apoptose , Infecções por Coxsackievirus/patologia , Camundongos , Miocardite/patologia , Miocardite/virologia , Poliubiquitina/imunologia
4.
Eur J Immunol ; 41(9): 2774-81, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21630249

RESUMO

Coxsackievirus B3 (CVB3)-infection is a frequent cause of acute myocarditis, which may result in chronic myocarditis and virus persistence. Investigation of the initial immune responses to CVB3 may shed light on the mechanisms that contribute to ongoing disease. DCs, as key professional APCs, were investigated in two MHC-matched hosts: while C57BL/6 mice are resistant to chronic CVB3-myocarditis, the A.BY/SnJ mouse strain exhibits susceptibility. DC maturation and activation were critically impaired in A.BY/SnJ mice, as reflected by the failure of DCs to induce co-stimulatory molecules and cytokine/chemokine responses. MHC class I-restricted antigen presentation via the cross-presentation pathway was also affected in DCs from A.BY/SnJ mice. DC maturation involves the accumulation of DC aggresome-like induced structures (DALISs) and the transient storage of defective ribosomal products (DRiPs). DCs from A.BY/SnJ mice showed permanent DALIS accumulation; the detection of poly-ubiquitinylated CVB3 proteins in these DALISs suggested a limitation in the MHC class I antigenic supply in this host. In conclusion, ongoing chronic disease in A.BY/SnJ mice due to a failure to clear the virus may be attributed to defects in DC maturation/activation and DC MHC class I antigen processing.


Assuntos
Infecções por Coxsackievirus/imunologia , Apresentação Cruzada , Células Dendríticas/metabolismo , Enterovirus/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Infecções por Coxsackievirus/complicações , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/patologia , Células Dendríticas/virologia , Enterovirus/patogenicidade , Antígenos de Histocompatibilidade Classe I/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miocardite/etiologia , Ubiquitinação , Virulência
5.
Am J Pathol ; 175(2): 510-8, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19590042

RESUMO

Murine models of coxsackievirus B3 (CVB3)-induced myocarditis mimic the divergent human disease course of cardiotropic viral infection, with host-specific outcomes ranging from complete recovery in resistant mice to chronic disease in susceptible hosts. To identify susceptibility factors that modulate the course of viral myocarditis, we show that type-I interferon (IFN) responses are considerably impaired in acute CVB3-induced myocarditis in susceptible mice, which have been linked to immunoproteasome (IP) formation. Here we report that in concurrence with distinctive type-I IFN kinetics, myocardial IP formation peaked early after infection in resistant mice and was postponed with maximum IP expression concomitant to massive inflammation and predominant type-II IFN responses in susceptible mice. IP activity is linked to a strong enhancement of antigenic viral peptide presentation. To investigate the impact of myocardial IPs in CVB3-induced myocarditis, we identified two novel CVB3 T cell epitopes, virus capsid protein 2 [285-293] and polymerase 3D [2170-2177]. Analysis of myocardial IPs in CVB3-induced myocarditis revealed that myocardial IP expression resulted in efficient epitope generation. As opposed to the susceptible host, myocardial IP expression at early stages of disease corresponded to enhanced CVB3 epitope generation in the hearts of resistant mice. We propose that this process may precondition the infected heart for adaptive immune responses. In conclusion, type-I IFN-induced myocardial IP activity at early stages coincides with less severe disease manifestation in CVB3-induced myocarditis.


Assuntos
Infecções por Enterovirus/imunologia , Enterovirus/imunologia , Interferon Tipo I/imunologia , Miocardite/imunologia , Miocardite/virologia , Complexo de Endopeptidases do Proteassoma/imunologia , Animais , Modelos Animais de Doenças , Infecções por Enterovirus/complicações , Infecções por Enterovirus/patologia , Epitopos de Linfócito T/imunologia , Humanos , Interferon Tipo I/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Miocardite/patologia , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...