Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Biol Cell ; 35(3): ar38, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38170572

RESUMO

The ubiquitination of transmembrane receptors regulates endocytosis, intracellular traffic, and signal transduction. Bone marrow-derived macrophages from myeloid Cbl-/- and Cbl-b-/- double knockout (DKO) mice display sustained proliferation mirroring the myeloproliferative disease that these mice succumb to. Here, we found that the ubiquitin ligases Cbl and Cbl-b have overlapping functions for controlling the endocytosis and intracellular traffic of the CSF-1R. DKO macrophages displayed complete loss of ubiquitination of the CSF-1R whereas partial ubiquitination was observed for either single Cbl-/- or Cbl-b-/- macrophages. Unlike wild type, DKO macrophages were immortal and displayed slower CSF-1R internalization, elevated AKT signaling, and a failure to transport the CSF-1R into the lumen of nascent macropinosomes, leaving its cytoplasmic region available for signaling. CSF-1R degradation depended upon lysosomal vATPase activity in both WT and DKO macrophages, with this degradation confined to macropinosomes in WT but occurring in distributed/tubular lysosomes in DKO cells. RNA-sequencing comparison of Cbl-/-, Cbl-b-/- and DKO macrophages indicated that while the overall macrophage transcriptional program remained intact, DKO macrophages had alterations in gene expression associated with growth factor signaling, cell cycle, inflammation and senescence. Cbl-b-/- had minimal effect on the transcriptional program whereas Cbl-/- led to more alternations but only DKO macrophages demonstrated substantial changes in the transcriptome, suggesting overlapping but unique functions for the two Cbl-family members. Thus, Cbl/Cbl-b-mediated ubiquitination of CSF-1R regulates its endocytic fate, constrains inflammatory gene expression, and regulates signaling for macrophage proliferation.


Assuntos
Receptor de Fator Estimulador de Colônias de Macrófagos , Ubiquitina , Camundongos , Animais , Ubiquitina/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Fator Estimulador de Colônias de Macrófagos/genética , Fator Estimulador de Colônias de Macrófagos/metabolismo , Fator Estimulador de Colônias de Macrófagos/farmacologia , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Macrófagos/metabolismo
2.
Nat Commun ; 12(1): 4838, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34376698

RESUMO

Macropinosomes are formed by shaping actin-rich plasma membrane ruffles into large intracellular organelles in a phosphatidylinositol 3-kinase (PI3K)-coordinated manner. Here, we utilize lattice lightsheet microscopy and image visualization methods to map the three-dimensional structure and dynamics of macropinosome formation relative to PI3K activity. We show that multiple ruffling morphologies produce macropinosomes and that the majority form through collisions of adjacent PI3K-rich ruffles. By combining multiple volumetric representations of the plasma membrane structure and PI3K products, we show that PI3K activity begins early throughout the entire ruffle volume and continues to increase until peak activity concentrates at the base of the ruffle after the macropinosome closes. Additionally, areas of the plasma membrane rich in ruffling had increased PI3K activity and produced many macropinosomes of various sizes. Pharmacologic inhibition of PI3K activity had little effect on the rate and morphology of membrane ruffling, demonstrating that early production of 3'-phosphoinositides within ruffles plays a minor role in regulating their morphology. However, 3'-phosphoinositides are critical for the fusogenic activity that seals ruffles into macropinosomes. Taken together, these data indicate that local PI3K activity is amplified in ruffles and serves as a priming mechanism for closure and sealing of ruffles into macropinosomes.


Assuntos
Membrana Celular/metabolismo , Microscopia de Fluorescência/métodos , Fosfatidilinositol 3-Quinases/metabolismo , Pinocitose/fisiologia , Animais , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Células HEK293 , Humanos , Macrófagos/citologia , Macrófagos/metabolismo , Macrófagos/ultraestrutura , Camundongos , Microscopia Eletrônica de Varredura , Morfolinas/farmacologia , Fosfatidilinositóis/metabolismo , Pinocitose/efeitos dos fármacos , Células RAW 264.7
3.
Front Immunol ; 11: 617767, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33679705

RESUMO

The inhibition of Fcγ receptors (FcγR) is an attractive strategy for treating diseases driven by IgG immune complexes (IC). Previously, we demonstrated that an engineered tri-valent arrangement of IgG1 Fc domains (SIF1) potently inhibited FcγR activation by IC, whereas a penta-valent Fc molecule (PentX) activated FcγR, potentially mimicking ICs and leading to Syk phosphorylation. Thus, a precise balance exists between the number of engaged FcγRs for inhibition versus activation. Here, we demonstrate that Fc valency differentially controls FcγR activation and inhibition within distinct subcellular compartments. Large Fc multimer clusters consisting of 5-50 Fc domains predominately recruited Syk-mScarlet to patches on the plasma membrane, whereas PentX exclusively recruited Syk-mScarlet to endosomes in human monocytic cell line (THP-1 cells). In contrast, SIF1, similar to monomeric Fc, spent longer periods docked to FcγRs on the plasma membrane and did not accumulate and recruit Syk-mScarlet within large endosomes. Single particle tracking (SPT) of fluorescent engineered Fc molecules and Syk-mScarlet at the plasma membrane imaged by total internal reflection fluorescence microscopy (SPT-TIRF), revealed that Syk-mScarlet sampled the plasma membrane was not recruited to FcγR docked with any of the engineered Fc molecules at the plasma membrane. Furthermore, the motions of FcγRs docked with recombinant Fc (rFc), SIF1 or PentX, displayed similar motions with D ~ 0.15 µm2/s, indicating that SIF1 and PentX did not induce reorganization or microclustering of FcγRs beyond the ligating valency. Multicolor SPT-TIRF and brightness analysis of docked rFc, SIF1 and PentX also indicated that FcγRs were not pre-assembled into clusters. Taken together, activation on the plasma membrane requires assembly of more than 5 FcγRs. Unlike rFc or SIF1, PentX accumulated Syk-mScarlet on endosomes indicating that the threshold for FcγR activation on endosomes is lower than on the plasma membrane. We conclude that the inhibitory effects of SIF1 are mediated by stabilizing a ligated and inactive FcγR on the plasma membrane. Thus, FcγR inhibition can be achieved by low valency ligation with SIF1 that behaves similarly to FcγR docked with monomeric IgG.


Assuntos
Fragmentos Fc das Imunoglobulinas/imunologia , Imunoglobulina G/imunologia , Fagocitose/imunologia , Receptores de IgG/metabolismo , Complexo Antígeno-Anticorpo/imunologia , Endossomos/imunologia , Humanos , Macrófagos/imunologia , Transdução de Sinais/imunologia
4.
Cell Signal ; 28(9): 1325-1335, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27224507

RESUMO

Colony stimulating factor-1 receptor (CSF-1R), a receptor tyrosine kinase (RTK), is the master regulator of macrophage biology. CSF-1 can bind CSF-1R resulting in receptor activation and signalling essential for macrophage functions such as proliferation, differentiation, survival, polarization, phagocytosis, cytokine secretion, and motility. CSF-1R activation can only occur after the receptor is presented on the macrophage cell surface. This process is reliant upon the underlying macrophage receptor trafficking machinery. However, the mechanistic details governing this process are incompletely understood. C-terminal Eps15 Homology Domain-containing (EHD) proteins have recently emerged as key regulators of receptor trafficking but have not yet been studied in the context of macrophage CSF-1R signalling. In this manuscript, we utilize primary bone-marrow derived macrophages (BMDMs) to reveal a novel function of EHD1 as a regulator of CSF-1R abundance on the cell surface. We report that EHD1-knockout (EHD1-KO) macrophages cell surface and total CSF-1R levels are significantly decreased. The decline in CSF-1R levels corresponds with reduced downstream macrophage functions such as cell proliferation, migration, and spreading. In EHD1-KO macrophages, transport of newly synthesized CSF-1R to the macrophage cell surface was reduced and was associated with the shunting of the receptor to the lysosome, which resulted in receptor degradation. These findings reveal a novel and functionally important role for EHD1 in governing CSF-1R signalling via regulation of anterograde transport of CSF-1R to the macrophage cell surface.


Assuntos
Membrana Celular/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Transdução de Sinais , Proteínas de Transporte Vesicular/metabolismo , Animais , Autoantígenos/metabolismo , Compartimento Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Macrolídeos/farmacologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Biossíntese de Proteínas/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
5.
BMC Immunol ; 17: 5, 2016 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-26970734

RESUMO

BACKGROUND: Recent evidence indicates that in addition to the T-cell receptor, microclustering is an important mechanism for the activation of the B-cell receptor and the mast cell Fcε-receptor. In macrophages and neutrophils, particles opsonized with immunoglobulin G (IgG) antibodies activate the phagocytic Fcγ-receptor (FcγR) leading to rearrangements of the actin cytoskeleton. The purpose of this study was to establish a system for high-resolution imaging of FcγR microclustering dynamics and the recruitment of the downstream signaling machinery to these microclusters. METHODS: We developed a supported lipid bilayer platform with incorporated antibodies on its surface to study the formation and maturation of FcγR signaling complexes in macrophages. Time-lapse multicolor total internal reflection microscopy was used to capture the formation of FcγR-IgG microclusters and their assembly into signaling complexes on the plasma membrane of murine bone marrow derived macrophages. RESULTS: Upon antibody binding, macrophages formed FcγR-IgG complexes at the leading edge of advancing pseudopods. These complexes then moved toward the center of the cell to form a structure reminiscent of the supramolecular complex observed in the T-cell/antigen presenting cell immune synapse. Colocalization of signaling protein Syk with nascent clusters of antibodies indicated that phosphorylated receptor complexes underwent maturation as they trafficked toward the center of the cell. Additionally, imaging of fluorescent BtkPH domains indicated that 3'-phosphoinositides propagated laterally away from the FcγR microclusters. CONCLUSION: We demonstrate that surface-associated but mobile IgG induces the formation of FcγR microclusters at the pseudopod leading edge. These clusters recruit Syk and drive the production of diffusing PI(3,4,5)P3 that is coordinated with lamellar actin polymerization. Upon reaching maximal extension, FcγR microclusters depart from the leading edge and are transported to the center of the cellular contact region to form a synapse-like structure, analogous to the process observed for T-cell receptors.


Assuntos
Imageamento Tridimensional , Macrófagos/metabolismo , Microscopia de Fluorescência/métodos , Fagocitose , Receptores de IgG/metabolismo , Transdução de Sinais , Actinas/metabolismo , Animais , Imunoglobulina G/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos Endogâmicos C57BL , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Tirosina Quinases/metabolismo , Quinase Syk
6.
J Cell Sci ; 127(Pt 24): 5228-39, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25335894

RESUMO

Activation of the macrophage colony stimulating factor-1 receptor (CSF-1R) by CSF-1 stimulates pronounced macropinocytosis and drives proliferation of macrophages. Although the role of macropinocytosis in CSF-1R signaling remains unknown, we show here that, despite internalizing large quantities of plasma membrane, macropinosomes contribute little to the internalization of the CSF-1-CSF-1R complex. Rather, internalization of the CSF-1R in small endocytic vesicles that are sensitive to clathrin disruption, outcompetes macropinosomes for CSF-1R endocytosis. Following internalization, small vesicles carrying the CSF-1R underwent homotypic fusion and then trafficked to newly formed macropinosomes bearing Rab5. As these macropinosomes matured, acquiring Rab7, the CSF-1R was transported into their lumen and degraded. Inhibition of macropinocytosis delayed receptor degradation despite no disruption to CSF-1R endocytosis. These data indicate that CSF-1-stimulated macropinosomes are sites of multivesicular body formation and accelerate CSF-1R degradation. Furthermore, we demonstrate that macropinocytosis and cell growth have a matching dose dependence on CSF-1, suggesting that macropinosomes might be a central mechanism coupling CSF-1R signaling and macrophage growth.


Assuntos
Macrófagos/metabolismo , Pinocitose , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Clatrina/metabolismo , Endocitose/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Humanos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Mitógenos/farmacologia , Modelos Biológicos , Pinocitose/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
7.
PLoS One ; 8(6): e64760, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23762252

RESUMO

Fluorescence Resonance Energy Transfer (FRET) microscopy has emerged as a powerful tool to visualize nanoscale protein-protein interactions while capturing their microscale organization and millisecond dynamics. Recently, FRET microscopy was extended to imaging of multiple donor-acceptor pairs, thereby enabling visualization of multiple biochemical events within a single living cell. These methods require numerous equations that must be defined on a case-by-case basis. Here, we present a universal multispectral microscopy method (N-Way FRET) to enable quantitative imaging for any number of interacting and non-interacting FRET pairs. This approach redefines linear unmixing to incorporate the excitation and emission couplings created by FRET, which cannot be accounted for in conventional linear unmixing. Experiments on a three-fluorophore system using blue, yellow and red fluorescent proteins validate the method in living cells. In addition, we propose a simple linear algebra scheme for error propagation from input data to estimate the uncertainty in the computed FRET images. We demonstrate the strength of this approach by monitoring the oligomerization of three FP-tagged HIV Gag proteins whose tight association in the viral capsid is readily observed. Replacement of one FP-Gag molecule with a lipid raft-targeted FP allowed direct observation of Gag oligomerization with no association between FP-Gag and raft-targeted FP. The N-Way FRET method provides a new toolbox for capturing multiple molecular processes with high spatial and temporal resolution in living cells.


Assuntos
Transferência Ressonante de Energia de Fluorescência/métodos , Proteínas do Vírus da Imunodeficiência Humana/química , Proteínas do Vírus da Imunodeficiência Humana/ultraestrutura , Microscopia Confocal/métodos , Proteínas de Bactérias , Capsídeo/química , Corantes Fluorescentes , Proteínas de Fluorescência Verde , HIV/metabolismo , HIV/ultraestrutura , Proteínas Luminescentes , Ligação Proteica , Mapas de Interação de Proteínas , Multimerização Proteica , Transdução de Sinais , Proteína Vermelha Fluorescente
8.
Microsc Microanal ; 19(2): 350-9, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23472941

RESUMO

Fluorescence resonance energy transfer (FRET) microscopy is a powerful technique to quantify dynamic protein-protein interactions in live cells. Total internal reflection fluorescence (TIRF) microscopy can selectively excite molecules within about 150 nm of the glass-cell interface. Recently, these two approaches were combined to enable high-resolution FRET imaging on the adherent surface of living cells. Here, we show that interference fringing of the coherent laser excitation used in TIRF creates lateral heterogeneities that impair quantitative TIRF-FRET measurements. We overcome this limitation by using a two-dimensional scan head to rotate laser beams for donor and acceptor excitation around the back focal plane of a high numerical aperture objective. By setting different radii for the circles traced out by each laser in the back focal plane, the penetration depth was corrected for different wavelengths. These modifications quell spatial variations in illumination and permit calibration for quantitative TIRF-FRET microscopy. The capability of TIRF-FRET was demonstrated by imaging assembled cyan and yellow fluorescent protein-tagged HIV-Gag molecules in single virions on the surfaces of living cells. These interactions are shown to be distinct from crowding of HIV-Gag in lipid rafts.


Assuntos
Transferência Ressonante de Energia de Fluorescência/métodos , HIV/metabolismo , Rim/virologia , Microscopia de Fluorescência/métodos , Vírion/metabolismo , Montagem de Vírus , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Células COS , Chlorocebus aethiops , Corantes Fluorescentes/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Rim/citologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética
9.
PLoS Pathog ; 9(2): e1003176, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23408893

RESUMO

Of the Orthomyxoviridae family of viruses, only influenza A viruses are thought to exist as multiple subtypes and has non-human maintenance hosts. In April 2011, nasal swabs were collected for virus isolation from pigs exhibiting influenza-like illness. Subsequent electron microscopic, biochemical, and genetic studies identified an orthomyxovirus with seven RNA segments exhibiting approximately 50% overall amino acid identity to human influenza C virus. Based on its genetic organizational similarities to influenza C viruses this virus has been provisionally designated C/Oklahoma/1334/2011 (C/OK). Phylogenetic analysis of the predicted viral proteins found that the divergence between C/OK and human influenza C viruses was similar to that observed between influenza A and B viruses. No cross reactivity was observed between C/OK and human influenza C viruses using hemagglutination inhibition (HI) assays. Additionally, screening of pig and human serum samples found that 9.5% and 1.3%, respectively, of individuals had measurable HI antibody titers to C/OK virus. C/OK virus was able to infect both ferrets and pigs and transmit to naive animals by direct contact. Cell culture studies showed that C/OK virus displayed a broader cellular tropism than a human influenza C virus. The observed difference in cellular tropism was further supported by structural analysis showing that hemagglutinin esterase (HE) proteins between two viruses have conserved enzymatic but divergent receptor-binding sites. These results suggest that C/OK virus represents a new subtype of influenza C viruses that currently circulates in pigs that has not been recognized previously. The presence of multiple subtypes of co-circulating influenza C viruses raises the possibility of reassortment and antigenic shift as mechanisms of influenza C virus evolution.


Assuntos
Anticorpos Antivirais/sangue , Gammainfluenzavirus/isolamento & purificação , Genoma Viral/genética , Infecções por Orthomyxoviridae/virologia , Doenças dos Suínos/virologia , Animais , Antígenos Virais/imunologia , Sequência de Bases , Técnicas de Cultura de Células , Furões , Testes de Inibição da Hemaglutinação , Hemaglutininas Virais/genética , Hemaglutininas Virais/metabolismo , Especificidade de Hospedeiro , Humanos , Gammainfluenzavirus/genética , Gammainfluenzavirus/imunologia , Gammainfluenzavirus/ultraestrutura , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Oklahoma , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/transmissão , Filogenia , Análise de Sequência de DNA , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/transmissão , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo
10.
Methods Mol Biol ; 827: 235-51, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22144279

RESUMO

Fluorescence resonance energy transfer (FRET) imaging can measure the spatial and temporal distributions of activated Rho GTPases within living cells. This information is essential for understanding how signaling networks influence Rho-GTPase switching and for elucidating the mechanisms of Rho GTPase control of the cytoskeleton. This chapter describes FRET microscopy methods to image the distribution of GTP-bound Rac and Cdc42 during the well-defined morphological transitions of phagocytosis by macrophages. Specifically, we describe the use of FRET microscopy to detect the binding of genetically encoded fluorescent protein fusions to Rac1 or Cdc42 with a fluorescent protein fusion to a p21-binding domain (PBD) that recognizes their GTP-bound states. We focus on quantifying the kinetics and activation levels of Rac and Cdc42 during Fc receptor-mediated phagocytosis by macrophages. This process is a Rac1, Cdc42, and actin-dependent process, by which macrophages engulf particles ranging in size from 0.5 to 20 µm and is an ideal model system for studying the spatial and temporal control of these GTPases. Quantitative FRET analysis for measuring the fractions of activated GTPase to allow comparison between cells, independent of the relative expression levels of the fluorescent fusions is also discussed.


Assuntos
Transferência Ressonante de Energia de Fluorescência/métodos , Macrófagos/metabolismo , Imagem Molecular/métodos , Fagocitose/fisiologia , Receptores Fc/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Animais , Células COS , Chlorocebus aethiops , Ativação Enzimática/fisiologia , Genes Reporter , Interpretação de Imagem Assistida por Computador , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Macrófagos/imunologia , Camundongos , Transfecção
11.
Proc Natl Acad Sci U S A ; 107(45): 19332-7, 2010 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-20974965

RESUMO

During Fcγ receptor (FcR)-mediated phagocytosis by macrophages, cytoplasm advances over IgG-coated particles by the sequential ligation of FcR in plasma membranes. If FcR signaling was strictly autonomous, then the signals generated during phagocytosis should be proportional to the number of ligated receptors. By measuring FcR-dependent responses to beads coated with various densities of IgG, this study identified nonlinear signaling that organizes an all or none response during particle ingestion. Phagocytosis of beads with IgG at low density either stalled after making small, actin-rich cups or proceeded to completion at the same rate as phagocytosis of high-density IgG beads. Signals were measured by quantifying the recruitment of YFP-labeled probes to phagocytic cup membranes. Although the magnitude of early signals correlated with IgG density, later signals showed an all or none response, which was regulated by the concentrations of 3' phosphoinositides in phagocytic cup membranes. Thus, 3' phosphoinositides, shown previously to be required for phagocytosis, function in a feedback regulatory mechanism affecting late but not early signals. This indicates a mechanism for the coordination of cell movements initiated by receptor signaling.


Assuntos
Macrófagos/imunologia , Fagocitose/imunologia , Receptores Fc/imunologia , Transdução de Sinais/imunologia , Animais , Linhagem Celular , Membrana Celular , Citoplasma , Retroalimentação Fisiológica , Imunoglobulina G , Camundongos , Fosfatidilinositóis/fisiologia
12.
Mol Biol Cell ; 21(3): 470-80, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19955216

RESUMO

Fcgamma Receptor (FcR)-mediated phagocytosis by macrophages requires phosphatidylinositol 3-kinase (PI3K) and activation of the Rho-family GTPases Cdc42 and Rac1. Cdc42 is activated at the advancing edge of the phagocytic cup, where actin is concentrated, and is deactivated at the base of the cup. The timing of 3' phosphoinositide (3'PI) concentration changes in cup membranes suggests a role for 3'PIs in deactivation of Cdc42. This study examined the relationships between PI3K and the patterns of Rho-family GTPase signaling during phagosome formation. Inhibition of PI3K resulted in persistently active Cdc42 and Rac1, but not Rac2, in stalled phagocytic cups. Patterns of 3'PIs and Rho-family GTPase activities during phagocytosis of 5- and 2-mum-diameter microspheres indicated similar underlying mechanisms despite particle size-dependent sensitivities to PI3K inhibition. Expression of constitutively active Cdc42(G12V) increased 3'PI concentrations in plasma membranes and small phagosomes, indicating a role for Cdc42 in PI3K activation. Cdc42(G12V) inhibited phagocytosis at a later stage than inhibition by dominant negative Cdc42(N17). Together, these studies identified a Cdc42 activation cycle organized by PI3K, in which FcR-activated Cdc42 stimulates PI3K and actin polymerization, and the subsequent increase of 3'PIs in cup membranes inactivates Cdc42 to allow actin recycling necessary for phagosome formation.


Assuntos
Fagocitose/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Receptores Fc/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Actinas/metabolismo , Animais , Ativação Enzimática , Inibidores Enzimáticos/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Microesferas , Tamanho da Partícula , Fagossomos/metabolismo , Fosfatidilinositol 3-Quinases/genética , Receptores Fc/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/fisiologia , Proteína cdc42 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
13.
J Cell Sci ; 122(Pt 18): 3250-61, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19690049

RESUMO

Macropinosomes are large endocytic vesicles that form in ruffling regions of plasma membrane. To analyze signal organization relative to ruffle closure into circular ruffles and cup closure into macropinosomes, this study used quantitative microscopy to measure 3' phosphoinositides and small-GTPase activities in a representative subset of forming macropinosomes. Macropinocytosis was stimulated by the addition of macrophage colony-stimulating factor (M-CSF) to macrophages expressing fluorescent reporter proteins. Ratiometric and fluorescence resonance energy transfer (FRET) microscopy determined that Rac1 activity and phosphatidylinositol (3,4,5)-trisphosphate [PtdIns(3,4,5)P(3)] levels increased transiently, peaking 26-30 seconds after ruffle closure. Three-dimensional reconstruction of cells labeled with the fluorescent dye FM4-64 showed that PtdIns(3,4,5)P(3) was restricted to open, circular cups in the plasma membrane. Quantitative fluorescence microscopic methods determined the timing of cup closure, which followed 40-100 seconds after Rac1 and PtdIns(3,4,5)P(3) deactivation and coincided with accumulation of phosphatidylinositol 3-phosphate and Rab5a. Thus, ruffle closure creates a circular domain of plasma membrane that localizes the activation and deactivation of Rac1 and phosphoinositide 3-kinase (PI3K), followed by recruitment of Rab5a and the contractile activities of cup closure.


Assuntos
Membrana Celular/metabolismo , Macrófagos/metabolismo , Pinocitose , Transdução de Sinais , Animais , Membrana Celular/enzimologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fatores de Tempo , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo
14.
Cell Microbiol ; 9(3): 790-803, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17140407

RESUMO

The signalling pathway for the hepatocyte growth factor receptor, Met/HGF-R, is hijacked by the bacterial surface protein InlB to induce Listeria monocytogenes entry into non-phagocytic cells. We previously showed that Listeria invades host cells by interacting with specialized microdomains of the host plasma membrane called lipid rafts. In this study, we analysed in living cells signalling events that are crucial for Listeria entry using a fluorescence resonance energy transfer-based microscopic method. Phosphoinositide (PI) 3-kinase activity and Rac1 signalling induced by Listeria interacting with epithelial cells were monitored as well as signalling induced by soluble InlB and the Met natural ligand HGF. We found that InlB and HGF induced similar kinetics of PI 3-kinase and Rac1 activation. PI 3-kinase activation was upstream and independent of Rac1 activation. Cholesterol-depletion experiments were performed to address the role of lipid rafts in Met signalling. The amount of 3'-phosphoinositides produced by PI 3-kinase was not affected by cholesterol depletion, while their membrane dynamic was cholesterol-dependent. Rac1 activation, downstream from PI 3-kinase, was cholesterol-dependent suggesting that the spatial distribution of 3'-phosphoinositides within membrane microdomains is critical for Rac1 activation and consequently for F-actin assembly at bacterial entry site.


Assuntos
Proteínas de Bactérias/fisiologia , Colesterol/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Proteínas de Membrana/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-met/fisiologia , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Chlorocebus aethiops , Colesterol/fisiologia , Cinética , Listeria monocytogenes/fisiologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microdomínios da Membrana/metabolismo , Microdomínios da Membrana/microbiologia , Microscopia de Fluorescência , Transfecção , Células Vero
15.
PLoS Biol ; 4(6): e162, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16669702

RESUMO

Fcgamma receptor (FcgammaR)-mediated phagocytosis of IgG-coated particles is regulated by 3'-phosphoinositides (3'PIs) and several classes of small GTPases, including ARF6 from the ADP Ribosylation Factor subfamily. The insensitivity of phagocytosis to brefeldin A (BFA), an inhibitor of certain ARF guanine nucleotide exchange factors (GEFs), previously indicated that ARF1 did not participate in phagocytosis. In this study, we show that ARF1 was activated during FcgammaR-mediated phagocytosis and that blocking normal ARF1 cycling inhibited phagosome closure. We examined the distributions and activation patterns of ARF6 and ARF1 during FcgammaR-mediated phagocytosis using fluorescence resonance energy transfer (FRET) stoichiometric microscopy of macrophages expressing CFP- or YFP-chimeras of ARF1, ARF6, and a GTP-ARF-binding protein domain. Both GTPases were activated by BFA-insensitive factors at sites of phagocytosis. ARF6 activation was restricted to the leading edge of the phagocytic cup, while ARF1 activation was delayed and delocalized over the phagosome. Phagocytic cups formed after inhibition of PI 3-kinase (PI-3K) contained persistently activated ARF6 and minimally activated ARF1. This indicates that a PI-3K-dependent signal transition defines the sequence of ARF GTPase activation during phagocytosis and that ARF6 and ARF1 coordinate different functions at the forming phagosome.


Assuntos
Fator 1 de Ribosilação do ADP/metabolismo , Fatores de Ribosilação do ADP/metabolismo , Fagocitose/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Receptores de IgG/fisiologia , Fator 1 de Ribosilação do ADP/análise , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/análise , Brefeldina A/farmacologia , Células Cultivadas , Ativação Enzimática , Transferência Ressonante de Energia de Fluorescência , Humanos , Macrófagos/metabolismo , Mutação , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais
16.
Cell Microbiol ; 8(5): 781-92, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16611227

RESUMO

Listeria monocytogenes (Lm) evade microbicidal defences inside macrophages by secreting a pore-forming cytolysin listeriolysin O (LLO), which allows Lm to escape vacuoles. LLO also inhibits Lm vacuole fusion with lysosomes, which indicates LLO alters vacuole chemistry prior to release of Lm into cytoplasm. Using fluorescent probes to measure membrane permeability, calcium and pH, we identified small membrane perforations in vacuoles containing wild-type but not LLO-deficient (hly-) Lm. The small membrane perforations released small fluorescent molecules and persisted for several minutes before expanding to allow exchange of larger fluorescent molecules. Macropinosomes and hly- Lm vacuoles acidified and increased their calcium content ([Ca2+]vac) within minutes of formation; however, the small perforations made by LLO-expressing bacteria increased vacuolar pH and decreased [Ca2+]vac shortly after infection. Experimental increases in vacuolar pH inhibited Lm vacuole fusion with lysosomes. The timing of perforation indicated that LLO-dependent delays of Lm vacuole maturation result from disruption of ion gradients across vacuolar membranes.


Assuntos
Cálcio/fisiologia , Membranas Intracelulares/microbiologia , Listeria monocytogenes/metabolismo , Vacúolos/fisiologia , Animais , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Linhagem Celular , Corantes Fluorescentes , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas , Concentração de Íons de Hidrogênio , Membranas Intracelulares/fisiologia , Listeria monocytogenes/genética , Lisossomos/fisiologia , Macrófagos/microbiologia , Macrófagos/fisiologia , Fusão de Membrana , Camundongos , Permeabilidade
17.
J Leukoc Biol ; 76(6): 1093-103, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15466916

RESUMO

Phagocytosis by macrophages can be initiated by Fcgamma receptors (FcR) in membranes that bind to Fc regions of immunoglobulin G (IgG). Activated FcR transduce signals to cytoplasm, which regulate the internalization of IgG-coated particles into plasma membrane-derived vacuoles, phagosomes. Particles internalized by phagocytosis are much larger than FcR, which prompts questions of if and how the receptors are coordinated with each other. FcR-mediated signal transduction entails recruitment of proteins from cytoplasm to the receptor, largely via protein phosphorylation. These FcR signaling complexes then activate proteins that regulate actin, myosin, membrane fusion, and the production of reactive oxygen intermediates. Recent fluorescence microscopic studies of phagocytosis in macrophages indicate that signaling by FcR occurs as a sequence of distinct stages, evident in the spatial and temporal patterns of phosphoinositides, protein kinase C, and Rho-family GTPase activation on forming phagosomes. The coordination of these stages may be regulated by lipids or lipid-anchored proteins, which diffuse away from FcR complexes. Lateral diffusion of FcR-derived signals could integrate FcR-dependent responses over large areas of membrane in the forming phagosome.


Assuntos
Macrófagos/imunologia , Fagocitose/imunologia , Receptores Fc/imunologia , Transdução de Sinais/imunologia , Animais , Membrana Celular/imunologia , Difusão , Ativação Enzimática/imunologia , Humanos , Imunoglobulina G/imunologia , Transporte Proteico/imunologia
18.
Mol Biol Cell ; 15(8): 3509-19, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15169870

RESUMO

The small G proteins Cdc42, Rac1, and Rac2 regulate the rearrangements of actin and membrane necessary for Fcgamma receptor-mediated phagocytosis by macrophages. Activated, GTP-bound Cdc42, Rac1, and Rac2 bind to the p21-binding domain (PBD) of PAK1, and this interaction provided a basis for microscopic methods to localize activation of these G proteins inside cells. Fluorescence resonance energy transfer-based stoichiometry of fluorescent chimeras of actin, PBD, Cdc42, Rac1, and Rac2 was used to quantify G protein activation relative to actin movements during phagocytosis of IgG-opsonized erythrocytes. The activation dynamics of endogenous G proteins, localized using yellow fluorescent protein-labeled PBD, was restricted to phagocytic cups, with a prominent spike of activation over an actin-poor region at the base of the cup. Refinements of fluorescence resonance energy transfer stoichiometry allowed calculation of the fractions of activated GTPases in forming phagosomes. Cdc42 activation was restricted to the leading margin of the cell, whereas Rac1 was active throughout the phagocytic cup. During phagosome closure, activation of Rac1 and Rac2 increased uniformly and transiently in the actin-poor region of phagosomal membrane. These distinct roles for Cdc42, Rac1, and Rac2 in the component activities of phagocytosis indicate mechanisms by which their differential regulation coordinates rearrangements of actin and membranes.


Assuntos
Macrófagos/enzimologia , Fagocitose/fisiologia , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Actinas/análise , Animais , Proteínas de Bactérias/análise , Linhagem Celular , Proteínas de Fluorescência Verde/análise , Proteínas Luminescentes/análise , Macrófagos/química , Macrófagos/metabolismo , Camundongos , Modelos Biológicos , Fagossomos/fisiologia , Fosfatidilinositol 3-Quinases/análise , Fosfatidilinositol 3-Quinases/metabolismo , Mapeamento de Interação de Proteínas , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Proteína cdc42 de Ligação ao GTP/análise , Quinases Ativadas por p21 , Proteínas rac de Ligação ao GTP/análise , Proteínas rac1 de Ligação ao GTP/análise , Proteína RAC2 de Ligação ao GTP
19.
J Cell Biol ; 164(2): 185-94, 2004 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-14718518

RESUMO

Many studies of endocytosis and phagocytosis presume that organelles containing a single kind of internalized particle exhibit invariant patterns of protein and phospholipid association as they mature inside cells. To test this presumption, fluorescent protein chimeras were expressed in RAW 264.7 macrophages, and time-lapse ratiometric fluorescence microscopy was used to measure the maturation dynamics of individual phagosomes containing IgG-opsonized erythrocytes. Quantitative analysis revealed consistent patterns of association for YFP chimeras of beta-actin, Rab5a, Rab7, and LAMP-1, and no association of YFP chimeras marking endoplasmic reticulum or Golgi. YFP-2xFYVE, recognizing phosphatidylinositol 3-phosphate (PI(3)P), showed two patterns of phagosome labeling. Some phagosomes increased labeling quickly after phagosome closure and then lost the label within 20 min, whereas others labeled more slowly and retained the label for several hours. The two patterns of PI(3)P on otherwise identical phagosomes indicated that organelle maturation does not necessarily follow a single path and that some features of phagosome maturation are integrated over the entire organelle.


Assuntos
Macrófagos/fisiologia , Fagossomos/fisiologia , Fagossomos/ultraestrutura , Animais , Linhagem Celular , Endocitose , Processamento de Imagem Assistida por Computador , Macrófagos/ultraestrutura , Camundongos , Microscopia de Fluorescência , Fagocitose
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA