Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Immunol Cell Biol ; 95(6): 564-570, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28290451

RESUMEN

Protein tyrosine phosphorylation guides many molecular interactions for cellular functions. SCIMP is a transmembrane adaptor protein (TRAP) family member that mediates selective proinflammatory cytokine responses generated by pathogen-activated Toll-like receptor (TLR) pathways in macrophages. TLR activation triggers SCIMP phosphorylation and selective phosphorylation of distinct tyrosine residues on this adaptor offers the potential for regulating or biasing inflammatory responses. To analyze site-specific phosphorylation events, we developed three probes based on the SH2 domains of known SCIMP effectors, and used them for pull-downs from macrophage extracts. CRISPR-mediated SCIMP-deficient RAW264.7 macrophage-like cells were reconstituted with various phosphorylation-deficient (Y58F, Y96F, Y120F) SCIMPs, and used to demonstrate the specificity of LPS/TLR4-induced, site-specific phosphorylation of SCIMP for the temporal recruitment of the effectors Grb2, Csk and SLP65. Our findings reveal potential for differential SCIMP phosphorylation and specific effectors to influence TLR signaling and inflammatory programs. Furthermore, the use of Csk-SH2 pull-downs to identify additional known and new Csk targets in LPS-activated macrophages reveals the wider utility of our SH2 probes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Interacciones Huésped-Patógeno , Dominios y Motivos de Interacción de Proteínas , Tirosina/metabolismo , Dominios Homologos src , Proteínas Adaptadoras Transductoras de Señales/química , Animales , Proteína Tirosina Quinasa CSK , Macrófagos/metabolismo , Ratones , Modelos Biológicos , Fosforilación , Unión Proteica , Células RAW 264.7 , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/metabolismo , Receptores Toll-Like/metabolismo , Familia-src Quinasas/metabolismo
2.
Immunol Cell Biol ; 92(3): 275-86, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24343664

RESUMEN

Rho GTPases are required for many cellular events such as adhesion, motility, and membrane trafficking. Here we show that in macrophages, the Rho GTPases Rac1 and Cdc42 are involved in lamellipodia and filopodia formation, respectively, and that both of these Rho GTPases are essential for the efficient surface delivery of tumor necrosis factor (TNF) to the plasma membrane following TLR4 stimulation. We have previously demonstrated intracellular trafficking of TNF via recycling endosomes in lipopolysaccharide (LPS)-activated macrophages. Here, we further define a specific role for Rac1 in intracellular TNF trafficking, demonstrating impairment in TNF release following TLR4 stimulation in the presence of a Rac inhibitor, in cells expressing a dominant negative (DN) form of Rac1, and following small interfering RNA (siRNA) knockdown of Rac1. Rac1 activity was required for TNF trafficking but not for TLR4 signaling following LPS stimulation. Reduced TNF secretion was due to a defect in Rac1 activity, but not of the closely related Rho GTPase Rac2, demonstrated by the additional use of macrophages derived from Rac2-deficient mice. Labeling recycling endosomes by the uptake of fluorescent transferrin enabled us to show that Rac1 was required for the final stages of TNF trafficking and delivery from recycling endosomes to the plasma membrane. Thus, actin remodeling by the Rho GTPase Rac1 is required for TNF cell surface delivery and release from macrophages.


Asunto(s)
Endocitosis , Endosomas/metabolismo , Macrófagos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Endocitosis/efectos de los fármacos , Endosomas/efectos de los fármacos , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/metabolismo , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Transporte de Proteínas/efectos de los fármacos , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Pironas/farmacología , Quinolinas/farmacología , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína RCA2 de Unión a GTP
3.
PLoS One ; 8(2): e57034, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23437303

RESUMEN

Lipopolysaccharide (LPS)-activated macrophages secrete pro-inflammatory cytokines, including tumor necrosis factor (TNF) to elicit innate immune responses. Secretion of these cytokines is also a major contributing factor in chronic inflammatory disease. In previous studies we have begun to elucidate the pathways and molecules that mediate the intracellular trafficking and secretion of TNF. Rab6a and Rab6a' (collectively Rab6) are trans-Golgi-localized GTPases known for roles in maintaining Golgi structure and Golgi-associated trafficking. We found that induction of TNF secretion by LPS promoted the selective increase of Rab6 expression. Depletion of Rab6 (via siRNA and shRNA) resulted in reorganization of the Golgi ribbon into more compact structures that at the resolution of electron microcopy consisted of elongated Golgi stacks that likely arose from fusion of smaller Golgi elements. Concomitantly, the delivery of TNF to the cell surface and subsequent release into the media was reduced. Dominant negative mutants of Rab6 had similar effects in disrupting TNF secretion. In live cells, Rab6-GFP were localized on trans-Golgi network (TGN)-derived tubular carriers demarked by the golgin p230. Rab6 depletion and inactive mutants altered carrier egress and partially reduced p230 membrane association. Our results show that Rab6 acts on TNF trafficking at the level of TGN exit in tubular carriers and our findings suggest Rab6 may stabilize p230 on the tubules to facilitate TNF transport. Both Rab6 isoforms are needed in macrophages for Golgi stack organization and for the efficient post-Golgi transport of TNF. This work provides new insights into Rab6 function and into the role of the Golgi complex in cytokine secretion in inflammatory macrophages.


Asunto(s)
Aparato de Golgi/metabolismo , Macrófagos/metabolismo , Factores de Necrosis Tumoral/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Autoantígenos/metabolismo , Brefeldino A/farmacología , Línea Celular , Membrana Celular/metabolismo , Proteínas de la Matriz de Golgi , Proteínas de la Membrana/metabolismo , Ratones , Mutación , Estabilidad Proteica , Transporte de Proteínas/efectos de los fármacos , Interferencia de ARN , Proteínas de Unión al GTP rab/genética
4.
J Biol Chem ; 280(11): 10478-83, 2005 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-15640147

RESUMEN

A key function of activated macrophages is to secrete proinflammatory cytokines such as TNFalpha; however, the intracellular pathway and machinery responsible for cytokine trafficking and secretion is largely undefined. Here we show that individual SNARE proteins involved in vesicle docking and fusion are regulated at both gene and protein expression upon stimulation with the bacterial cell wall component lipopolysaccharide. Focusing on two intracellular SNARE proteins, Vti1b and syntaxin 6 (Stx6), we show that they are up-regulated in conjunction with increasing cytokine secretion in activated macrophages and that their levels are selectively titrated to accommodate the volume and timing of post-Golgi cytokine trafficking. In macrophages, Vti1b and syntaxin 6 are localized on intracellular membranes and are present on isolated Golgi membranes and on Golgi-derived TNFalpha vesicles budded in vitro. By immunoprecipitation, we find that Vti1b and syntaxin 6 interact to form a novel intracellular Q-SNARE complex. Functional studies using overexpression of full-length and truncated proteins show that both Vti1b and syntaxin 6 function and have rate-limiting roles in TNFalpha trafficking and secretion. This study shows how macrophages have uniquely adapted a novel Golgi-associated SNARE complex to accommodate their requirement for increased cytokine secretion.


Asunto(s)
Proteínas Portadoras/metabolismo , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba , Animales , Proteínas Portadoras/química , Línea Celular , Citocinas/metabolismo , Exocitosis , Aparato de Golgi/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Immunoblotting , Inmunoprecipitación , Inflamación , Lipopolisacáridos/metabolismo , Proteínas de la Membrana/química , Ratones , Microscopía Fluorescente , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Qa-SNARE , Proteínas Qb-SNARE , ARN/metabolismo , Proteínas SNARE , Factores de Tiempo , Proteínas de Transporte Vesicular/metabolismo
5.
Traffic ; 4(3): 175-89, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12656990

RESUMEN

Galpha interacting protein (GAIP) is a regulator of G protein signaling protein that associates dynamically with vesicles and has been implicated in membrane trafficking, although its specific role is not yet known. Using an in vitro budding assay, we show that GAIP is recruited to a specific population of trans-Golgi network-derived vesicles and that these are distinct from coatomer or clathrin-coated vesicles. A truncation mutant (NT-GAIP) encoding only the N-terminal half of GAIP is recruited to trans-Golgi network membranes during the formation of vesicle carriers. Overexpression of NT-GAIP induces the formation of long, coated tubules, which are stabilized by microtubules. Results from the budding assay and from imaging in live cells show that these tubules remain attached to the Golgi stack rather than being released as carrier vesicles. NT-GAIP expression blocks membrane budding and results in the accumulation of tubular carrier intermediates. NT-GAIP-decorated tubules are competent to load vesicular stomatitis virus protein G-green fluorescent protein as post-Golgi, exocytic cargo and in cells expressing NT-GAIP there is reduced surface delivery of vesicular stomatitis virus protein G-green fluorescent protein. We conclude that GAIP functions as an essential part of the membrane budding machinery for a subset of post-Golgi exocytic carriers derived from the trans-Golgi network.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Red trans-Golgi/fisiología , Red trans-Golgi/ultraestructura , Animales , Fraccionamiento Celular , Proteínas Activadoras de GTPasa/genética , Proteínas Fluorescentes Verdes , Células HeLa , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía por Video , Mutagénesis , Conejos , Proteínas Recombinantes de Fusión/metabolismo , Eliminación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA