Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Eur J Cancer ; 70: 62-74, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27886573

RESUMO

Epidermal growth factor receptor (EGFR) plays a central role in the progression of several human malignancies. Although EGFR is a membrane receptor, it undergoes nuclear translocation, where it has a distinct signalling pathway. Herein, we report a novel mechanism by which cancer cells can directly transport EGFR to the nucleus of other cells via extracellular vesicles (EVs). The transported receptor is active and stimulates the nuclear EGFR pathways. Interestingly, the translocation of EGFR via EVs occurs independently of the nuclear localisation sequence that is required for nuclear translocation of endogenous EGFR. Also, we found that the mutant receptor EGFRvIII could be transported to the nucleus of other cells via EVs. To assess the role of EVs in the regulation of an actual nuclear receptor, we studied the regulation of androgen receptor (AR). We found that full-length AR and mutant variant ARv7 are secreted in EVs derived from prostate cancer cell lines and could be transported to the nucleus of AR-null cells. The EV-derived AR was able to bind the androgen-responsive promoter region of prostate specific antigen, and recruit RNA Pol II, an indication of active transcription. The nuclear-translocated AR via EVs enhanced the proliferation of acceptor cells in the absence of androgen. Finally, we provide evidence that nuclear localisation of AR could occur in vivo via orthotopically-injected EVs in male SCID mice prostate glands. To our knowledge, this is the first study showing the nuclear translocation of nuclear receptors via EVs, which significantly extends the role of EVs as paracrine transcriptional regulators.


Assuntos
Núcleo Celular/metabolismo , Receptores ErbB/metabolismo , Vesículas Extracelulares/metabolismo , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos SCID
3.
Methods Mol Biol ; 1458: 195-202, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27581023

RESUMO

Extracellular vesicles (ECV) are membrane compartments shed from all types of cells in various physiological and pathological states. In recent years, ECV have gained an increasing interest from the scientific community for their role as an intercellular communicator that plays important roles in modifying the tumor microenvironment. Multiple techniques have been established to collect ECV from conditioned media of cell culture or physiological fluids. The gold standard methodology is differential centrifugation. Although alternative techniques exist to collect ECV, these techniques have not proven suitable as a substitution for the ultracentrifugation procedure.


Assuntos
Vesículas Extracelulares/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Microambiente Tumoral , Transporte Biológico , Técnicas de Cultura de Células , Fracionamento Celular/métodos , Linhagem Celular , Micropartículas Derivadas de Células/metabolismo , Exossomos , Humanos , Ultracentrifugação/métodos
4.
PLoS Pathog ; 11(6): e1004962, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26061049

RESUMO

Pore formation is the most energy-demanding step during virus-induced membrane fusion, where high curvature of the fusion pore rim increases the spacing between lipid headgroups, exposing the hydrophobic interior of the membrane to water. How protein fusogens breach this thermodynamic barrier to pore formation is unclear. We identified a novel fusion-inducing lipid packing sensor (FLiPS) in the cytosolic endodomain of the baboon reovirus p15 fusion-associated small transmembrane (FAST) protein that is essential for pore formation during cell-cell fusion and syncytiogenesis. NMR spectroscopy and mutational studies indicate the dependence of this FLiPS on a hydrophobic helix-loop-helix structure. Biochemical and biophysical assays reveal the p15 FLiPS preferentially partitions into membranes with high positive curvature, and this partitioning is impeded by bis-ANS, a small molecule that inserts into hydrophobic defects in membranes. Most notably, the p15 FLiPS can be functionally replaced by heterologous amphipathic lipid packing sensors (ALPS) but not by other membrane-interactive amphipathic helices. Furthermore, a previously unrecognized amphipathic helix in the cytosolic domain of the reptilian reovirus p14 FAST protein can functionally replace the p15 FLiPS, and is itself replaceable by a heterologous ALPS motif. Anchored near the cytoplasmic leaflet by the FAST protein transmembrane domain, the FLiPS is perfectly positioned to insert into hydrophobic defects that begin to appear in the highly curved rim of nascent fusion pores, thereby lowering the energy barrier to stable pore formation.


Assuntos
Fusão de Membrana/fisiologia , Reoviridae/fisiologia , Proteínas Virais de Fusão/metabolismo , Animais , Chlorocebus aethiops , Cromatografia Líquida de Alta Pressão , Dicroísmo Circular , Espectroscopia de Ressonância Magnética , Conformação Proteica , Reoviridae/patogenicidade , Transfecção , Células Vero , Proteínas Virais de Fusão/química
5.
J Gen Virol ; 94(Pt 5): 1039-1050, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23343626

RESUMO

Piscine reovirus (PRV) is a tentative new member of the family Reoviridae and has been linked to heart and skeletal muscle inflammation in farmed Atlantic salmon (Salmo salar L.). Recent sequence-based evidence suggests that PRV is about equally related to members of the genera Orthoreovirus and Aquareovirus. Sequence similarities have also suggested that PRV might encode a fusion-associated small transmembrane (FAST) protein, which in turn suggests that PRV might be the prototype of a new genus with syncytium-inducing potential. In previous support of this designation has been the absence of identifiable PRV-encoded homologues of either the virion outer-clamp protein of ortho- and aquareoviruses or the virion outer-fibre protein of most orthoreoviruses. In the current report, we have provided experimental evidence that the putative p13 FAST protein of PRV lacks the defining feature of the FAST protein family - the ability to induce syncytium formation. Instead, p13 is the first example of a cytosolic, integral membrane protein encoded by ortho- or aquareoviruses, and induces cytotoxicity in the absence of cell-cell fusion. Sequence analysis also identified signature motifs of the outer-clamp and outer-fibre proteins of other reoviruses in two of the predicted PRV gene products. Based on these findings, we conclude that PRV does not encode a FAST protein and is therefore unlikely to be a new fusogenic reovirus. The presence of a novel integral membrane protein and two previously unrecognized, essential outer-capsid proteins has important implications for the biology, evolution and taxonomic classification of this virus.


Assuntos
Proteínas do Capsídeo/genética , Doenças dos Peixes/virologia , Proteínas de Membrana/genética , Infecções por Reoviridae/veterinária , Reoviridae/classificação , Salmão , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas do Capsídeo/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Citoplasma , Células Gigantes , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Orthoreovirus/classificação , Orthoreovirus/genética , Orthoreovirus/isolamento & purificação , Orthoreovirus/metabolismo , Filogenia , Proteínas Recombinantes de Fusão , Reoviridae/química , Reoviridae/genética , Reoviridae/isolamento & purificação , Infecções por Reoviridae/virologia , Alinhamento de Sequência , Células Vero , Vírion
6.
J Biol Chem ; 287(5): 3403-14, 2012 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-22170056

RESUMO

The p15 fusion-associated small transmembrane (FAST) protein is a nonstructural viral protein that induces cell-cell fusion and syncytium formation. The exceptionally small, myristoylated N-terminal ectodomain of p15 lacks any of the defining features of a typical viral fusion protein. NMR and CD spectroscopy indicate this small fusion module comprises a left-handed polyproline type II (PPII) helix flanked by small, unstructured N and C termini. Individual prolines in the 6-residue proline-rich motif are highly tolerant of alanine substitutions, but multiple substitutions that disrupt the PPII helix eliminate cell-cell fusion activity. A synthetic p15 ectodomain peptide induces lipid mixing between liposomes, but with unusual kinetics that involve a long lag phase before the onset of rapid lipid mixing, and the length of the lag phase correlates with the kinetics of peptide-induced liposome aggregation. Lipid mixing, liposome aggregation, and stable peptide-membrane interactions are all dependent on both the N-terminal myristate and the presence of the PPII helix. We present a model for the mechanism of action of this novel viral fusion peptide, whereby the N-terminal myristate mediates initial, reversible peptide-membrane binding that is stabilized by subsequent amino acid-membrane interactions. These interactions induce a biphasic membrane fusion reaction, with peptide-induced liposome aggregation representing a distinct, rate-limiting event that precedes membrane merger. Although the prolines in the proline-rich motif do not directly interact with membranes, the PPII helix may function to force solvent exposure of hydrophobic amino acid side chains in the regions flanking the helix to promote membrane binding, apposition, and fusion.


Assuntos
Lipoilação , Modelos Químicos , Ácido Mirístico/química , Peptídeos/química , Reoviridae/química , Proteínas Virais de Fusão/química , Motivos de Aminoácidos , Animais , Chlorocebus aethiops , Lipossomos/química , Lipossomos/metabolismo , Ácido Mirístico/metabolismo , Ressonância Magnética Nuclear Biomolecular , Peptídeos/síntese química , Peptídeos/genética , Peptídeos/metabolismo , Estrutura Terciária de Proteína , Reoviridae/genética , Reoviridae/metabolismo , Células Vero , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo
7.
Methods ; 55(2): 122-6, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21958986

RESUMO

Membrane fusion is a protein catalyzed biophysical reaction that involves the simultaneous intermixing of two phospholipid bilayers and of the aqueous compartments bound by their respective bilayers. In the case of enveloped virus fusogens, short hydrophobic or amphipathic fusion peptides that are components of the larger fusion complex are essential for the membrane merger event. The process of cell-cell membrane fusion and syncytium formation induced by the nonenveloped fusogenic orthoreoviruses is driven by the Fusion-Associated Small Transmembrane (FAST) proteins, which are similarly dependent on the action of fusion peptides. In this article, we describe some simple methods for the biophysical characterization of viral membrane fusion peptides. Liposomes serve as an ideal model system for characterizing peptide-membrane interactions because their size, shape and composition can be readily manipulated. We present details of fluorescence assays used to elucidate the kinetics of membrane fusion as well as complimentary assays used to characterize peptide-induced liposome binding and aggregation.


Assuntos
Proteínas Virais de Fusão/química , Internalização do Vírus , Animais , Linhagem Celular Tumoral , Núcleo Celular , Emulsões/química , Transferência Ressonante de Energia de Fluorescência , Fluorometria/instrumentação , Fluorometria/métodos , Células Gigantes , Lipossomos/química , Proteínas de Fusão de Membrana , Fosfolipídeos/química , Ligação Proteica , Rodaminas/química , Sonicação/métodos , Espectrofotometria/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA