Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
mBio ; 9(4)2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-30042202

RESUMO

To transfer the viral genome into the host cell cytoplasm, internalized influenza A virus (IAV) particles depend on the fusion of the IAV envelope with host endosomal membranes. The antiviral host interferon (IFN) response includes the upregulation of interferon-induced transmembrane protein 3 (IFITM3), which inhibits the release of the viral content into the cytosol. Although IFITM3 induction occurs concomitantly with late endosomal/lysosomal (LE/L) cholesterol accumulation, the functional significance of this process is not well understood. Here we report that LE/L cholesterol accumulation itself plays a pivotal role in the early antiviral defense. We demonstrate that inducing LE/L cholesterol accumulation is antiviral in non-IFN-primed cells, restricting incoming IAV particles and impairing mixing of IAV/endosomal membrane lipids. Our results establish a protective function of LE/L cholesterol accumulation and suggest endosomal cholesterol balance as a possible antiviral target.IMPORTANCE With annual epidemics occurring in all parts of the world and the risk of global outbreaks, influenza A virus (IAV) infections remain a major threat to public health. Infected host cells detect viral components and mount an interferon (IFN)-mediated response to restrict virus propagation and spread of infection. Identification of cellular factors and underlying mechanisms that establish such an antiviral state can provide novel strategies for the development of antiviral drugs. The contribution of LE/L cholesterol levels, especially in the context of the IFN-induced antiviral response, has remained controversial so far. Here, we report that accumulation of cholesterol in the LE/L compartment contributes to the IFN-induced host cell defense against incoming IAV. Our results establish cholesterol accumulation in LE/L per se as a novel antiviral barrier and suggest the endosomal cholesterol balance as a putative druggable host cell factor in IAV infection.


Assuntos
Colesterol/metabolismo , Endossomos/metabolismo , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Vírus da Influenza A/fisiologia , Células A549 , Anexina A6/genética , Linhagem Celular , Endossomos/virologia , Humanos , Interferons/imunologia , Lisossomos/metabolismo , Lisossomos/virologia , Proteínas de Membrana/genética , Proteínas de Ligação a RNA/genética , Regulação para Cima , Replicação Viral
2.
Biol Chem ; 397(10): 949-59, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27366904

RESUMO

Emerging infectious diseases and drug-resistant infectious agents call for the development of innovative antimicrobial strategies. With pathogenicity now considered to arise from the complex and bi-directional interplay between a microbe and the host, host cell factor targeting has emerged as a promising approach that might overcome the limitations of classical antimicrobial drug development and could open up novel and efficient therapeutic strategies. Interaction with and modulation of host cell membranes is a recurrent theme in the host-microbe relationship. In this review, we provide an overview of what is currently known about the role of the Ca2+ dependent, membrane-binding annexin protein family in pathogen-host interactions, and discuss their emerging functions as host cell derived auxiliary proteins in microbe-host interactions and host cell targets.


Assuntos
Anexinas/metabolismo , Interações Hospedeiro-Patógeno , Animais , Humanos , Microbiologia , Terapia de Alvo Molecular
3.
mBio ; 4(6): e00608-13, 2013 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-24194536

RESUMO

UNLABELLED: Influenza is caused by influenza A virus (IAV), an enveloped, negative-stranded RNA virus that derives its envelope lipids from the host cell plasma membrane. Here, we examined the functional role of cellular cholesterol in the IAV infection cycle. We show that shifting of cellular cholesterol pools via the Ca(2+)-regulated membrane-binding protein annexin A6 (AnxA6) affects the infectivity of progeny virus particles. Elevated levels of cellular AnxA6, which decrease plasma membrane and increase late endosomal cholesterol levels, impaired IAV replication and propagation, whereas RNA interference-mediated AnxA6 ablation increased viral progeny titers. Pharmacological accumulation of late endosomal cholesterol also diminished IAV virus propagation. Decreased IAV replication caused by upregulated AnxA6 expression could be restored either by exogenous replenishment of host cell cholesterol or by ectopic expression of the late endosomal cholesterol transporter Niemann-Pick C1 (NPC1). Virus released from AnxA6-overexpressing cells displayed significantly reduced cholesterol levels. Our results show that IAV replication depends on maintenance of the cellular cholesterol balance and identify AnxA6 as a critical factor in linking IAV to cellular cholesterol homeostasis. IMPORTANCE: Influenza A virus (IAV) is a major public health concern, and yet, major host-pathogen interactions regulating IAV replication still remain poorly understood. It is known that host cell cholesterol is a critical factor in the influenza virus life cycle. The viral envelope is derived from the host cell membrane during the process of budding and, hence, equips the virus with a special lipid-protein mixture which is high in cholesterol. However, the influence of host cell cholesterol homeostasis on IAV infection is largely unknown. We show that IAV infection success critically depends on host cell cholesterol distribution. Cholesterol sequestration in the endosomal compartment impairs progeny titer and infectivity and is associated with reduced cholesterol content in the viral envelope.


Assuntos
Anexina A6/metabolismo , Membrana Celular/metabolismo , Colesterol/metabolismo , Endossomos/metabolismo , Interações Hospedeiro-Patógeno , Vírus da Influenza A/fisiologia , Replicação Viral , Anexina A6/genética , Linhagem Celular , Membrana Celular/química , Endossomos/química , Células Epiteliais/química , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Vírus da Influenza A/química , Carga Viral , Vírion/química
4.
Biochim Biophys Acta ; 1833(7): 1700-11, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23246849

RESUMO

Endocytosis of activated growth factor receptors regulates spatio-temporal cellular signaling. In the case of the EGF receptor, sorting into multivesicular bodies (MVBs) controls signal termination and subsequently leads to receptor degradation in lysosomes. Annexin A1, a Ca(2+)-regulated membrane binding protein often deregulated in human cancers, interacts with the EGF receptor and is phosphorylated by internalized EGF receptor on endosomes. Most relevant for EGF receptor signal termination, annexin A1 is required for the formation of internal vesicles in MVBs that sequester ligand-bound EGF receptor away from the limiting membrane. To elucidate the mechanism underlying annexin A1-dependent EGF receptor trafficking we employed an N-terminally truncated annexin A1 mutant that lacks the EGF receptor phosphorylation site and the site for interaction with its protein ligand S100A11. Overexpression of this dominant-negative mutant induces a delay in EGF-induced EGF receptor transport to the LAMP1-positive late endosomal/lysosomal compartment and impairs ligand-induced EGF receptor degradation. Consistent with these findings, EGF-stimulated EGF receptor and MAP kinase pathway signaling is prolonged. Importantly, depletion of S100A11 also results in a delayed EGF receptor transport and prolonged MAP kinase signaling comparable to the trafficking defect observed in cells expressing the N-terminally truncated annexin A1 mutant. These results strongly suggest that the function of annexin A1 as a regulator of EGF receptor trafficking, degradation and signaling is critically mediated through an N-terminal interaction with S100A11 in the endosomal compartment. This interaction appears to be essential for lysosomal targeting of the EGF receptor, possibly by providing a physical scaffold supporting inward vesiculation in MVBs. This article is part of a Special Issue entitled: 12th European Symposium on Calcium.


Assuntos
Anexina A1/metabolismo , Movimento Celular , Proliferação de Células , Receptores ErbB/metabolismo , Proteínas de Membrana Lisossomal/metabolismo , Proteínas S100/metabolismo , Anexina A1/antagonistas & inibidores , Anexina A1/genética , Compartimento Celular , Ensaio de Unidades Formadoras de Colônias , Endocitose/fisiologia , Endossomos/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/genética , Células HeLa , Humanos , Técnicas Imunoenzimáticas , Proteínas de Membrana Lisossomal/genética , Lisossomos/metabolismo , Microscopia Confocal , Microscopia de Fluorescência , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Transporte Proteico , Proteínas S100/antagonistas & inibidores , Proteínas S100/genética , Ressonância de Plasmônio de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA