Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Nat Med ; 13(6): 703-10, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17496899

RESUMO

Cerebral malaria claims more than 1 million lives per year. We report that heme oxygenase-1 (HO-1, encoded by Hmox1) prevents the development of experimental cerebral malaria (ECM). BALB/c mice infected with Plasmodium berghei ANKA upregulated HO-1 expression and activity and did not develop ECM. Deletion of Hmox1 and inhibition of HO activity increased ECM incidence to 83% and 78%, respectively. HO-1 upregulation was lower in infected C57BL/6 compared to BALB/c mice, and all infected C57BL/6 mice developed ECM (100% incidence). Pharmacological induction of HO-1 and exposure to the end-product of HO-1 activity, carbon monoxide (CO), reduced ECM incidence in C57BL/6 mice to 10% and 0%, respectively. Whereas neither HO-1 nor CO affected parasitemia, both prevented blood-brain barrier (BBB) disruption, brain microvasculature congestion and neuroinflammation, including CD8(+) T-cell brain sequestration. These effects were mediated by the binding of CO to hemoglobin, preventing hemoglobin oxidation and the generation of free heme, a molecule that triggers ECM pathogenesis.


Assuntos
Monóxido de Carbono/fisiologia , Heme Oxigenase-1/fisiologia , Heme/metabolismo , Malária Cerebral/enzimologia , Animais , Modelos Animais de Doenças , Heme Oxigenase-1/deficiência , Heme Oxigenase-1/genética , Malária Cerebral/tratamento farmacológico , Malária Cerebral/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Plasmodium berghei
2.
PLoS Pathog ; 7(12): e1002422, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22174682

RESUMO

Autophagy is a conserved degradative pathway used as a host defense mechanism against intracellular pathogens. However, several viruses can evade or subvert autophagy to insure their own replication. Nevertheless, the molecular details of viral interaction with autophagy remain largely unknown. We have determined the ability of 83 proteins of several families of RNA viruses (Paramyxoviridae, Flaviviridae, Orthomyxoviridae, Retroviridae and Togaviridae), to interact with 44 human autophagy-associated proteins using yeast two-hybrid and bioinformatic analysis. We found that the autophagy network is highly targeted by RNA viruses. Although central to autophagy, targeted proteins have also a high number of connections with proteins of other cellular functions. Interestingly, immunity-associated GTPase family M (IRGM), the most targeted protein, was found to interact with the autophagy-associated proteins ATG5, ATG10, MAP1CL3C and SH3GLB1. Strikingly, reduction of IRGM expression using small interfering RNA impairs both Measles virus (MeV), Hepatitis C virus (HCV) and human immunodeficiency virus-1 (HIV-1)-induced autophagy and viral particle production. Moreover we found that the expression of IRGM-interacting MeV-C, HCV-NS3 or HIV-NEF proteins per se is sufficient to induce autophagy, through an IRGM dependent pathway. Our work reveals an unexpected role of IRGM in virus-induced autophagy and suggests that several different families of RNA viruses may use common strategies to manipulate autophagy to improve viral infectivity.


Assuntos
Autofagia/fisiologia , Proteínas de Ligação ao GTP/metabolismo , Infecções por Vírus de RNA/metabolismo , Infecções por Vírus de RNA/transmissão , Vírus de RNA/metabolismo , Sequência de Bases , Western Blotting , Biologia Computacional , Proteínas de Ligação ao GTP/genética , Células HeLa , Humanos , Microscopia Confocal , Dados de Sequência Molecular , Fases de Leitura Aberta/genética , Infecções por Vírus de RNA/genética , Vírus de RNA/genética , RNA Interferente Pequeno , Transfecção , Técnicas do Sistema de Duplo-Híbrido , Proteínas Virais/metabolismo
3.
Autophagy ; 8(7): 1136-7, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22722598

RESUMO

Several intracellular pathogens have the ability to avoid or exploit the otherwise destructive process of autophagy. RNA viruses are constantly confronted with cellular autophagy, and several of them hijack autophagy during the infectious cycle to improve their own replication. Nevertheless, our knowledge of viral molecular strategies used to manipulate autophagy remains limited. Our study allowed the identification of molecular interactions between 44 autophagy-associated proteins and 83 viral proteins belonging to five different RNA virus families. This interactome revealed that the autophagy network machinery is highly targeted by RNA viruses. Interestingly, whereas some autophagy-associated proteins are targeted by only one RNA virus family, others are recurrent targets of several families. Among them, we found IRGM as the most targeted autophagy-associated protein. Downregulation of IRGM expression prevents autophagy induction by measles virus, HCV and HIV-1, and compromises viral replication. Our work combined interactomic and analytical approaches to identify potential pathogen virulence factors targeting autophagy.

4.
Autophagy ; 6(2): 299-300, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20087059

RESUMO

Autophagy is a degradative mechanism involved in cell protection against invading pathogens. Although the autophagic process is well characterized, the molecular pathways leading to its activation upon pathogen binding remain poorly understood. Our recent work demonstrates that the cell surface pathogen receptor CD46 induces autophagy upon pathogen recognition. The molecular pathway linking CD46 to the autophagosome machinery relies on the scaffold protein GOPC and on the autophagosome formation complex Beclin 1/VPS34. The CD46-dependent autophagy is critical to an early control of infection.


Assuntos
Autofagia/imunologia , Proteína Cofatora de Membrana/imunologia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Interações Hospedeiro-Patógeno , Camundongos , Fagossomos/metabolismo
5.
Cell Host Microbe ; 6(4): 354-66, 2009 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-19837375

RESUMO

Autophagy is a highly regulated self-degradative mechanism required at a basal level for intracellular clearance and recycling of cytoplasmic contents. Upon intracellular pathogen invasion, autophagy can be induced as an innate immune mechanism to control infection. Nevertheless, pathogens have developed strategies to avoid or hijack autophagy for their own benefit. The molecular pathways inducing autophagy in response to infection remain poorly documented. We report here that the engagement of CD46, a ubiquitous human surface receptor able to bind several different pathogens, is sufficient to induce autophagy. CD46-Cyt-1, one of the two C-terminal splice variants of CD46, is linked to the autophagosome formation complex VPS34/Beclin1 via its interaction with the scaffold protein GOPC. Measles virus and group A Streptococcus, two CD46-binding pathogens, induce autophagy through a CD46-Cyt-1/GOPC pathway. Thus, upon microorganism recognition, a cell surface pathogen receptor can directly trigger autophagy, a critical step to control infection.


Assuntos
Autofagia , Vírus do Sarampo/imunologia , Proteína Cofatora de Membrana/imunologia , Streptococcus pyogenes/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Proteínas de Transporte/metabolismo , Proteínas da Matriz do Complexo de Golgi , Humanos , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Fosfatidilinositol 3-Quinases/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas
6.
J Immunol ; 179(11): 7840-51, 2007 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18025230

RESUMO

Heme oxygenase-1 (HO-1; encoded by the Hmox1 gene) catalyzes the degradation of free heme into biliverdin, via a reaction that releases iron (Fe) and carbon monoxide. We report that HO-1 down-regulates the proinflammatory phenotype associated with endothelial cell (EC) activation by reducing intracellular nonprotein-bound Fe (labile Fe). EC isolated from Hmox1(-/-) mice have higher levels of intracellular labile Fe and reactive oxygen species (ROS) as compared with EC isolated from Hmox1(+/+) mice. Basal and TNF-induced expression of VCAM-1, ICAM-1, and E-selectin were increased in Hmox1(-/-) vs Hmox1(+/+) EC, an effect reversed by Fe chelation using deferoxamine mesylate (DFO). Fe chelation inhibits TNF-driven transcription of Vcam-1, Icam-1, and E-selectin, as assessed using luciferase reporter assays. This effect is associated with inhibition of the transcription factor NF-kappaB via a mechanism that is not associated with the inhibition of IkappaBalpha phosphorylation/degradation or NF-kappaB (i.e., RelA) nuclear translocation, although it affects very modestly NF-kappaB binding to DNA kappaB consensus sequences in the Vcam-1 and E-selectin promoters. HO-1 inhibits NF-kappaB (i.e., RelA) phosphorylation at Ser(276), a phosphoacceptor that is critical to sustain TNF-driven NF-kappaB activity in EC. This effect was mimicked by Fe chelation as well as by antioxidants (N-acetylcysteine). In conclusion, we demonstrate a novel mechanism via which HO-1 down-modulates the proinflammatory phenotype of activated EC, i.e., the inhibition of RelA phosphorylation at Ser(276).


Assuntos
Moléculas de Adesão Celular/imunologia , Células Endoteliais/imunologia , Regulação da Expressão Gênica/imunologia , Heme Oxigenase-1/imunologia , Serina/imunologia , Fator de Transcrição RelA/metabolismo , Animais , Moléculas de Adesão Celular/efeitos dos fármacos , Moléculas de Adesão Celular/genética , Células Cultivadas , Regulação para Baixo/imunologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Compostos Férricos/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/deficiência , Heme Oxigenase-1/farmacologia , Inflamação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , NF-kappa B/antagonistas & inibidores , NF-kappa B/imunologia , Fosforilação/efeitos dos fármacos , Serina/efeitos dos fármacos , Fator de Transcrição RelA/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Molécula 1 de Adesão de Célula Vascular/biossíntese , Molécula 1 de Adesão de Célula Vascular/efeitos dos fármacos
7.
J Immunol ; 177(3): 1894-903, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16849502

RESUMO

Heme oxygenase-1 (HO-1) protects endothelial cells (EC) from undergoing apoptosis. This effect is mimicked by CO, generated via the catabolism of heme by HO-1. The antiapoptotic effect of CO in EC was abrogated when activation of the p38alpha and p38beta MAPKs was inhibited by the pyridinyl imidazole SB202190. Using small interfering RNA, p38beta was found to be cytoprotective in EC, whereas p38alpha was not. When overexpressed in EC, HO-1 targeted specifically the p38alpha but not the p38beta MAPK isoform for degradation by the 26S proteasome, an effect reversed by the 26S proteasome inhibitors MG-132 or lactacystin. Inhibition of p38alpha expression was also observed when HO-1 was induced physiologically by iron protoporphyrin IX (hemin). Inhibition of p38alpha no longer occurred when HO activity was inhibited by tin protoporphyrin IX, suggesting that p38alpha degradation was mediated by an end product of heme catabolism. Exogenous CO inhibited p38alpha expression in EC, suggesting that CO is the end product that mediates this effect. The antiapoptotic effect of HO-1 was impaired when p38alpha expression was restored ectopically or when its degradation by the 26S proteasome was inhibited by MG-132. Furthermore, the antiapoptotic effect of HO-1 was lost when p38beta expression was targeted by a specific p38beta small interfering RNA. In conclusion, the antiapoptotic effect of HO-1 in EC is dependent on the degradation of p38alpha by the 26S proteasome and on the expression of p38beta.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Células Endoteliais/enzimologia , Endotélio Vascular/citologia , Endotélio Vascular/enzimologia , Heme Oxigenase-1/fisiologia , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Animais , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Monóxido de Carbono/fisiologia , Bovinos , Linhagem Celular , Citoproteção/fisiologia , Células Endoteliais/citologia , Ativação Enzimática/fisiologia , Células HeLa , Heme Oxigenase-1/antagonistas & inibidores , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/biossíntese , Isoenzimas/genética , Isoenzimas/metabolismo , Proteína Quinase 11 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 11 Ativada por Mitógeno/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 14 Ativada por Mitógeno/biossíntese , Proteína Quinase 14 Ativada por Mitógeno/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Complexo de Endopeptidases do Proteassoma/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Transfecção
8.
J Immunol ; 172(6): 3553-63, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15004156

RESUMO

Heme oxygenase-1 (HO-1) cleaves the porphyrin ring of heme into carbon monoxide, Fe2+, and biliverdin, which is then converted into bilirubin. Heme-derived Fe2+ induces the expression of the iron-sequestering protein ferritin and activates the ATPase Fe2+-secreting pump, which decrease intracellular free Fe2+ content. Based on the antioxidant effect of bilirubin and that of decreased free cellular Fe2+, we questioned whether HO-1 would modulate the expression of proinflammatory genes associated with endothelial cell (EC) activation. We tested this hypothesis specifically for the genes E-selectin (CD62), ICAM-1 (CD54), and VCAM-1 (CD106). We found that HO-1 overexpression in EC inhibited TNF-alpha-mediated E-selectin and VCAM-1, but not ICAM-1 expression, as tested at the RNA and protein level. Heme-driven HO-1 expression had similar effects to those of overexpressed HO-1. In addition, HO-1 inhibited the activation of NF-kappaB, a transcription factor required for TNF-alpha-mediated up-regulation of these genes in EC. Bilirubin and/or Fe2+ chelation mimicked the effects of HO-1, whereas biliverdin or carbon monoxide did not. In conclusion, HO-1 inhibits the expression of proinflammatory genes associated with EC activation via a mechanism that is associated with the inhibition of NF-kappaB activation. This effect of HO-1 is mediated by bilirubin and/or by a decrease of free intracellular Fe2+ but probably not by biliverdin or carbon monoxide.


Assuntos
Endotélio Vascular/enzimologia , Endotélio Vascular/imunologia , Heme Oxigenase (Desciclizante)/fisiologia , Adenoviridae/genética , Animais , Bilirrubina/farmacologia , Monóxido de Carbono/farmacologia , Bovinos , Moléculas de Adesão Celular , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Selectina E/biossíntese , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Heme/farmacologia , Heme Oxigenase (Desciclizante)/biossíntese , Heme Oxigenase (Desciclizante)/genética , Heme Oxigenase-1 , Humanos , Molécula 1 de Adesão Intercelular/biossíntese , Quelantes de Ferro/farmacologia , Sistema de Sinalização das MAP Quinases/imunologia , Proteínas de Membrana , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/fisiologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/farmacologia , Suínos , Transdução Genética , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/fisiologia , Regulação para Cima/imunologia , Molécula 1 de Adesão de Célula Vascular/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA