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.
Biochim Biophys Acta ; 1863(6 Pt A): 1106-18, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26862060

RESUMO

Pseudomonas aeruginosa is an opportunistic pathogen that induces severe lung infections such as ventilator-associated pneumonia and acute lung injury. Under these conditions, the bacterium diminishes epithelial integrity and inhibits tissue repair mechanisms, leading to persistent infections. Understanding the involved bacterial virulence factors and their mode of action is essential for the development of new therapeutic approaches. In our study we discovered a so far unknown effect of the P. aeruginosa lectin LecB on host cell physiology. LecB alone was sufficient to attenuate migration and proliferation of human lung epithelial cells and to induce transcriptional activity of NF-κB. These effects are characteristic of impaired tissue repair. Moreover, we found a strong degradation of ß-catenin, which was partially recovered by the proteasome inhibitor lactacystin. In addition, LecB induced loss of cell-cell contacts and reduced expression of the ß-catenin targets c-myc and cyclin D1. Blocking of LecB binding to host cell plasma membrane receptors by soluble l-fucose prevented these changes in host cell behavior and signaling, and thereby provides a powerful strategy to suppress LecB function. Our findings suggest that P. aeruginosa employs LecB as a virulence factor to induce ß-catenin degradation, which then represses processes that are directly linked to tissue recovery.


Assuntos
Proteínas de Bactérias/farmacologia , Células Epiteliais/efeitos dos fármacos , Lectinas/farmacologia , beta Catenina/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Proteínas de Bactérias/genética , Western Blotting , Comunicação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Inibidores de Cisteína Proteinase/farmacologia , Relação Dose-Resposta a Droga , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Integrina beta1/metabolismo , Lectinas/genética , Microscopia Confocal , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Fator de Transcrição RelA/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos
2.
Biochim Biophys Acta ; 1860(2): 392-401, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26546712

RESUMO

BACKGROUND: Fruiting body lectins have been proposed to act as effector proteins in the defense of fungi against parasites and predators. The Marasmius oreades agglutinin (MOA) is a lectin from the fairy ring mushroom with specificity for Galα1-3Gal containing carbohydrates. This lectin is composed of an N-terminal carbohydrate-binding domain and a C-terminal dimerization domain. The dimerization domain of MOA shows in addition calcium-dependent cysteine protease activity, similar to the calpain family. METHODS: Cell detachment assay, cell viability assay, immunofluorescence, live cell imaging and Western blot using MDCKII cell line. RESULTS: In this study, we demonstrate in MDCKII cells that after internalization, MOA protease activity induces profound physiological cellular responses, like cytoskeleton rearrangement, cell detachment and cell death. These changes are preceded by a decrease in FAK phosphorylation and an internalization and degradation of ß1-integrin, consistent with a disruption of integrin-dependent cell adhesion signaling. Once internalized, MOA accumulates in late endosomal compartments. CONCLUSION: Our results suggest a possible toxic mechanism of MOA, which consists of disturbing the cell adhesion and the cell viability. GENERAL SIGNIFICANCE: After being ingested by a predator, MOA might exert a protective role by diminishing host cell integrity.


Assuntos
Aglutininas/fisiologia , Integrina beta1/fisiologia , Marasmius/química , Animais , Adesão Celular , Células Cultivadas , Clatrina/fisiologia , Cães , Dinaminas/fisiologia , Endocitose , Endossomos/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/fisiologia
3.
Mol Biosyst ; 12(2): 345-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26672971

RESUMO

One major regulatory mechanism in cell signalling is the spatio-temporal control of the localization of signalling molecules. We synthetically designed an entire cell signalling pathway, which allows controlling the transport of signalling molecules from the plasma membrane to the nucleus, by using light and small molecules.


Assuntos
Proteínas de Arabidopsis/fisiologia , Núcleo Celular/metabolismo , Fitocromo B/fisiologia , Transdução de Sinais/efeitos da radiação , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Endopeptidases/fisiologia , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Células HeLa , Humanos , Engenharia Metabólica , Transfecção
4.
ACS Synth Biol ; 4(9): 951-8, 2015 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-25803699

RESUMO

Protein trafficking in and out of the nucleus represents a key step in controlling cell fate and function. Here we report the development of a red light-inducible and far-red light-reversible synthetic system for controlling nuclear localization of proteins in mammalian cells and zebrafish. First, we synthetically reconstructed and validated the red light-dependent Arabidopsis phytochrome B nuclear import mediated by phytochrome-interacting factor 3 in a nonplant environment and support current hypotheses on the import mechanism in planta. On the basis of this principle we next regulated nuclear import and activity of target proteins by the spatiotemporal projection of light patterns. A synthetic transcription factor was translocated into the nucleus of mammalian cells and zebrafish to drive transgene expression. These data demonstrate the first in vivo application of a plant phytochrome-based optogenetic tool in vertebrates and expand the repertoire of available light-regulated molecular devices.


Assuntos
Transporte Ativo do Núcleo Celular/efeitos da radiação , Núcleo Celular/efeitos da radiação , Engenharia Genética , Luz , Animais , Arabidopsis/metabolismo , Proteínas de Arabidopsis/metabolismo , Células CHO , Cricetulus , Camundongos , Células NIH 3T3 , Fitocromo B/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Peixe-Zebra
5.
Proc Natl Acad Sci U S A ; 111(35): 12895-900, 2014 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-25136128

RESUMO

Glycosphingolipids are important structural constituents of cellular membranes. They are involved in the formation of nanodomains ("lipid rafts"), which serve as important signaling platforms. Invasive bacterial pathogens exploit these signaling domains to trigger actin polymerization for the bending of the plasma membrane and the engulfment of the bacterium--a key process in bacterial uptake. However, it is unknown whether glycosphingolipids directly take part in the membrane invagination process. Here, we demonstrate that a "lipid zipper," which is formed by the interaction between the bacterial surface lectin LecA and its cellular receptor, the glycosphingolipid Gb3, triggers plasma membrane bending during host cell invasion of the bacterium Pseudomonas aeruginosa. In vitro experiments with Gb3-containing giant unilamellar vesicles revealed that LecA/Gb3-mediated lipid zippering was sufficient to achieve complete membrane engulfment of the bacterium. In addition, theoretical modeling elucidated that the adhesion energy of the LecA-Gb3 interaction is adequate to drive the engulfment process. In cellulo experiments demonstrated that inhibition of the LecA/Gb3 lipid zipper by either lecA knockout, Gb3 depletion, or application of soluble sugars that interfere with LecA binding to Gb3 significantly lowered P. aeruginosa uptake by host cells. Of note, membrane engulfment of P. aeruginosa occurred independently of actin polymerization, thus corroborating that lipid zippering alone is sufficient for this crucial first step of bacterial host-cell entry. Our study sheds new light on the impact of glycosphingolipids in the cellular invasion of bacterial pathogens and provides a mechanistic explication of the initial uptake processes.


Assuntos
Actinas/metabolismo , Glicoesfingolipídeos/metabolismo , Microdomínios da Membrana/microbiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/metabolismo , Adesinas Bacterianas/metabolismo , Aderência Bacteriana/fisiologia , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Glicolipídeos/metabolismo , Bicamadas Lipídicas/metabolismo , Microdomínios da Membrana/metabolismo , Modelos Biológicos , Transdução de Sinais/fisiologia , Esfingolipídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...