Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Elife ; 122023 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-37796108

RESUMEN

The T cell receptor (TCR) is a complex molecular machine that directs the activation of T cells, allowing the immune system to fight pathogens and cancer cells. Despite decades of investigation, the molecular mechanism of TCR activation is still controversial. One of the leading activation hypotheses is the allosteric model. This model posits that binding of pMHC at the extracellular domain triggers a dynamic change in the transmembrane (TM) domain of the TCR subunits, which leads to signaling at the cytoplasmic side. We sought to test this hypothesis by creating a TM ligand for TCR. Previously we described a method to create a soluble peptide capable of inserting into membranes and binding to the TM domain of the receptor tyrosine kinase EphA2 (Alves et al., eLife, 2018). Here, we show that the approach is generalizable to complex membrane receptors, by designing a TM ligand for TCR. We observed that the designed peptide caused a reduction of Lck phosphorylation of TCR at the CD3ζ subunit in T cells. As a result, in the presence of this peptide inhibitor of TCR (PITCR), the proximal signaling cascade downstream of TCR activation was significantly dampened. Co-localization and co-immunoprecipitation in diisobutylene maleic acid (DIBMA) native nanodiscs confirmed that PITCR was able to bind to the TCR. AlphaFold-Multimer predicted that PITCR binds to the TM region of TCR, where it interacts with the two CD3ζ subunits. Our results additionally indicate that PITCR disrupts the allosteric changes in the compactness of the TM bundle that occur upon TCR activation, lending support to the allosteric TCR activation model. The TCR inhibition achieved by PITCR might be useful to treat inflammatory and autoimmune diseases and to prevent organ transplant rejection, as in these conditions aberrant activation of TCR contributes to disease.


Asunto(s)
Receptores de Antígenos de Linfocitos T , Linfocitos T , Ligandos , Receptores de Antígenos de Linfocitos T/metabolismo , Fosforilación , Péptidos/farmacología , Péptidos/metabolismo
2.
Small ; 19(20): e2207805, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36811150

RESUMEN

Photosynthetic light harvesting requires efficient energy transfer within dynamic networks of light-harvesting complexes embedded within phospholipid membranes. Artificial light-harvesting models are valuable tools for understanding the structural features underpinning energy absorption and transfer within chromophore arrays. Here, a method for attaching a protein-based light-harvesting model to a planar, fluid supported lipid bilayer (SLB) is developed.  The protein model consists of the tobacco mosaic viral capsid proteins that are gene-doubled to create a tandem dimer (dTMV). Assemblies of dTMV break the facial symmetry of the double disk to allow for differentiation between the disk faces. A single reactive lysine residue is incorporated into the dTMV assemblies for the site-selective attachment of chromophores for light absorption. On the opposing dTMV face, a cysteine residue is incorporated for the bioconjugation of a peptide containing a polyhistidine tag for association with SLBs. The dual-modified dTMV complexes show significant association with SLBs and exhibit mobility on the bilayer. The techniques used herein offer a new method for protein-surface attachment and provide a platform for evaluating excited state energy transfer events in a dynamic, fully synthetic artificial light-harvesting system.


Asunto(s)
Fotosíntesis , Proteínas , Transferencia de Energía , Membrana Dobles de Lípidos/química
3.
Proc Natl Acad Sci U S A ; 119(19): e2122531119, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35507881

RESUMEN

We reconstitute a phosphotyrosine-mediated protein condensation phase transition of the ∼200 residue cytoplasmic tail of the epidermal growth factor receptor (EGFR) and the adaptor protein, Grb2, on a membrane surface. The phase transition depends on phosphorylation of the EGFR tail, which recruits Grb2, and crosslinking through a Grb2-Grb2 binding interface. The Grb2 Y160 residue plays a structurally critical role in the Grb2-Grb2 interaction, and phosphorylation or mutation of Y160 prevents EGFR:Grb2 condensation. By extending the reconstitution experiment to include the guanine nucleotide exchange factor, SOS, and its substrate Ras, we further find that the condensation state of the EGFR tail controls the ability of SOS, recruited via Grb2, to activate Ras. These results identify an EGFR:Grb2 protein condensation phase transition as a regulator of signal propagation from EGFR to the MAPK pathway.


Asunto(s)
Receptores ErbB , Transducción de Señal , Receptores ErbB/metabolismo , Proteína Adaptadora GRB2/metabolismo , Fosforilación , Fosfotirosina/metabolismo
4.
Biophys J ; 121(10): 1897-1908, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35430415

RESUMEN

Cells sense a variety of extracellular growth factors and signaling molecules through numerous distinct receptor tyrosine kinases (RTKs) on the cell surface. In many cases, the same intracellular signaling molecules interact with more than one type of RTK. How signals from different RTKs retain the identity of the triggering receptor and how (or if) different receptors may synergize or compete remain largely unknown. Here we utilize an experimental strategy, combining microscale patterning and single-molecule imaging, to measure the competition between ephrin-A1:EphA2 and epidermal growth factor (EGF):EGF receptor (EGFR) ligand-receptor complexes for the shared downstream signaling molecules, Grb2 and SOS. The results reveal a distinct hierarchy, in which newly formed EGF:EGFR complexes outcompete ephrin-A1:EphA2 for Grb2 and SOS, revealing a type of negative crosstalk interaction fundamentally controlled by chemical mass action and protein copy number limitations.


Asunto(s)
Efrina-A1 , Receptor EphA2 , Factor de Crecimiento Epidérmico , Receptores ErbB/metabolismo , Retroalimentación , Receptor EphA2/metabolismo , Transducción de Señal
5.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33653954

RESUMEN

Ras dimerization is critical for Raf activation. Here we show that the Ras binding domain of Raf (Raf-RBD) induces robust Ras dimerization at low surface densities on supported lipid bilayers and, to a lesser extent, in solution as observed by size exclusion chromatography and confirmed by SAXS. Community network analysis based on molecular dynamics simulations shows robust allosteric connections linking the two Raf-RBD D113 residues located in the Galectin scaffold protein binding site of each Raf-RBD molecule and 85 Å apart on opposite ends of the dimer complex. Our results suggest that Raf-RBD binding and Ras dimerization are concerted events that lead to a high-affinity signaling complex at the membrane that we propose is an essential unit in the macromolecular assembly of higher order Ras/Raf/Galectin complexes important for signaling through the Ras/Raf/MEK/ERK pathway.


Asunto(s)
Simulación de Dinámica Molecular , Proteínas Proto-Oncogénicas p21(ras)/química , Quinasas raf/química , Galectinas/química , Galectinas/genética , Galectinas/metabolismo , Humanos , Dominios Proteicos , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Quinasas raf/genética , Quinasas raf/metabolismo
8.
Commun Biol ; 3(1): 429, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32764731

RESUMEN

The Eph family of receptor tyrosine kinases is crucial for assembly and maintenance of healthy tissues. Dysfunction in Eph signaling is causally associated with cancer progression. In breast cancer cells, dysregulated Eph signaling has been linked to alterations in receptor clustering abilities. Here, we implemented a single-cell assay and a scoring scheme to systematically probe the spatial organization of activated EphA receptors in multiple carcinoma cells. We show that cancer cells retain EphA clustering phenotype over several generations, and the degree of clustering reported for migration potential both at population and single-cell levels. Finally, using patient-derived cancer lines, we probed the evolution of EphA signalling in cell populations that underwent metastatic transformation and acquisition of drug resistance. Taken together, our scalable approach provides a reliable scoring scheme for EphA clustering that is consistent over multiple carcinomas and can assay heterogeneity of cancer cell populations in a cost- and time-effective manner.


Asunto(s)
Carcinoma/genética , Familia de Multigenes/genética , Receptores de la Familia Eph/genética , Análisis de la Célula Individual , Carcinoma/patología , Heterogeneidad Genética , Humanos , Fenotipo , Transducción de Señal/genética
9.
Proc Natl Acad Sci U S A ; 116(30): 15013-15022, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31278151

RESUMEN

Phosphorylation reactions, driven by competing kinases and phosphatases, are central elements of cellular signal transduction. We reconstituted a native eukaryotic lipid kinase-phosphatase reaction that drives the interconversion of phosphatidylinositol-4-phosphate [PI(4)P] and phosphatidylinositol-4,5-phosphate [PI(4,5)P2] on membrane surfaces. This system exhibited bistability and formed spatial composition patterns on supported membranes. In smaller confined regions of membrane, rapid diffusion ensures the system remains spatially homogeneous, but the final outcome-a predominantly PI(4)P or PI(4,5)P2 membrane composition-was governed by the size of the reaction environment. In larger confined regions, interplay between the reactions, diffusion, and confinement created a variety of differentially patterned states, including polarization. Experiments and kinetic modeling reveal how these geometric confinement effects arise from a mechanism based on stochastic fluctuations in the copy number of membrane-bound kinases and phosphatases. The underlying requirements for such behavior are unexpectedly simple and likely to occur in natural biological signaling systems.


Asunto(s)
Proteínas Bacterianas/química , Factores de Intercambio de Guanina Nucleótido/química , Fosfatidilinositol 4,5-Difosfato/química , Fosfatos de Fosfatidilinositol/química , Monoéster Fosfórico Hidrolasas/química , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Proteínas Bacterianas/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Cinética , Legionella pneumophila/química , Legionella pneumophila/enzimología , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosfolipasa C delta/química , Fosfolipasa C delta/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transducción de Señal , Imagen Individual de Molécula , Procesos Estocásticos , Liposomas Unilamelares/química , Liposomas Unilamelares/metabolismo
10.
Proc Natl Acad Sci U S A ; 116(22): 10798-10803, 2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-31076553

RESUMEN

The transformation of molecular binding events into cellular decisions is the basis of most biological signal transduction. A fundamental challenge faced by these systems is that reliance on protein-ligand chemical affinities alone generally results in poor sensitivity to ligand concentration, endangering the system to error. Here, we examine the lipid-binding pleckstrin homology and Tec homology (PH-TH) module of Bruton's tyrosine kinase (Btk). Using fluorescence correlation spectroscopy (FCS) and membrane-binding kinetic measurements, we identify a phosphatidylinositol (3-5)-trisphosphate (PIP3) sensing mechanism that achieves switch-like sensitivity to PIP3 levels, surpassing the intrinsic affinity discrimination of PIP3:PH binding. This mechanism employs multiple PIP3 binding as well as dimerization of Btk on the membrane surface. Studies in live cells confirm that mutations at the dimer interface and peripheral site produce effects comparable to that of the kinase-dead Btk in vivo. These results demonstrate how a single protein module can institute an allosteric counting mechanism to achieve high-precision discrimination of ligand concentration. Furthermore, this activation mechanism distinguishes Btk from other Tec family member kinases, Tec and Itk, which we show are not capable of dimerization through their PH-TH modules. This suggests that Btk plays a critical role in the stringency of the B cell response, whereas T cells rely on other mechanisms to achieve stringency.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/química , Agammaglobulinemia Tirosina Quinasa/metabolismo , Transducción de Señal/fisiología , Animales , Linfocitos B/metabolismo , Línea Celular , Pollos , Ratones , Modelos Moleculares , Mutación , Fosfatos de Fosfatidilinositol/metabolismo , Fosforilación , Conformación Proteica , Dominios Proteicos/fisiología , Multimerización de Proteína
11.
Sci Signal ; 12(564)2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30647147

RESUMEN

T cell receptor (TCR) binding to agonist peptide major histocompatibility complex (pMHC) triggers signaling events that initiate T cell responses. This system is remarkably sensitive, requiring only a few binding events to successfully activate a cellular response. On average, activating pMHC ligands exhibit mean dwell times of at least a few seconds when bound to the TCR. However, a T cell accumulates pMHC-TCR interactions as a stochastic series of discrete, single-molecule binding events whose individual dwell times are broadly distributed. With activation occurring in response to only a handful of such binding events, individual cells are unlikely to experience the average binding time. Here, we mapped the ensemble of pMHC-TCR binding events in space and time while simultaneously monitoring cellular activation. Our findings revealed that T cell activation hinges on rare, long-dwell time binding events that are an order of magnitude longer than the average agonist pMHC-TCR dwell time. Furthermore, we observed that short pMHC-TCR binding events that were spatially correlated and temporally sequential led to cellular activation. These observations indicate that T cell antigen discrimination likely occurs by sensing the tail end of the pMHC-TCR binding dwell time distribution rather than its average properties.


Asunto(s)
Activación de Linfocitos/inmunología , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Algoritmos , Secuencia de Aminoácidos , Animales , Células Cultivadas , Cinética , Ligandos , Complejo Mayor de Histocompatibilidad/inmunología , Ratones Transgénicos , Microscopía Fluorescente , Péptidos/química , Péptidos/metabolismo , Unión Proteica , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo
12.
Biophys J ; 115(5): 865-873, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30075851

RESUMEN

Interactions between EphB4 receptor tyrosine kinases and their membrane-bound ephrin-B2 ligands on apposed cells play a regulatory role in neural stem cell differentiation. With both receptor and ligand constrained to move within the membranes of their respective cells, this signaling system inevitably experiences spatial confinement and mechanical forces in conjunction with receptor-ligand binding. In this study, we reconstitute the EphB4-ephrin-B2 juxtacrine signaling geometry using a supported-lipid-bilayer system presenting laterally mobile and monomeric ephrin-B2 ligands to live neural stem cells. This experimental platform successfully reconstitutes EphB4-ephrin-B2 binding, lateral clustering, downstream signaling activation, and neuronal differentiation, all in a configuration that preserves the spatiomechanical aspects of the natural juxtacrine signaling geometry. Additionally, the supported bilayer system allows control of lateral movement and clustering of the receptor-ligand complexes through patterns of physical barriers to lateral diffusion fabricated onto the underlying substrate. The results from this study reveal a distinct spatiomechanical effect on the ability of EphB4-ephrin-B2 signaling to induce neuronal differentiation. These observations parallel similar studies of the EphA2-ephrin-A1 system in a very different biological context, suggesting that such spatiomechanical regulation may be a common feature of Eph-ephrin signaling.


Asunto(s)
Diferenciación Celular , Efrina-B2/metabolismo , Fenómenos Mecánicos , Células-Madre Neurales/citología , Receptor EphB4/metabolismo , Transducción de Señal , Animales , Fenómenos Biomecánicos , Membrana Celular/metabolismo , Ratones
13.
Cell ; 174(3): 672-687.e27, 2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-30053426

RESUMEN

TCR-signaling strength generally correlates with peptide-MHC binding affinity; however, exceptions exist. We find high-affinity, yet non-stimulatory, interactions occur with high frequency in the human T cell repertoire. Here, we studied human TCRs that are refractory to activation by pMHC ligands despite robust binding. Analysis of 3D affinity, 2D dwell time, and crystal structures of stimulatory versus non-stimulatory TCR-pMHC interactions failed to account for their different signaling outcomes. Using yeast pMHC display, we identified peptide agonists of a formerly non-responsive TCR. Single-molecule force measurements demonstrated the emergence of catch bonds in the activating TCR-pMHC interactions, correlating with exclusion of CD45 from the TCR-APC contact site. Molecular dynamics simulations of TCR-pMHC disengagement distinguished agonist from non-agonist ligands based on the acquisition of catch bonds within the TCR-pMHC interface. The isolation of catch bonds as a parameter mediating the coupling of TCR binding and signaling has important implications for TCR and antigen engineering for immunotherapy.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/fisiología , Activación de Linfocitos/fisiología , Adulto , Femenino , Humanos , Cinética , Ligandos , Complejo Mayor de Histocompatibilidad/fisiología , Masculino , Persona de Mediana Edad , Simulación de Dinámica Molecular , Oligopéptidos , Péptidos , Unión Proteica/fisiología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal , Imagen Individual de Molécula , Linfocitos T/fisiología
14.
Proc Natl Acad Sci U S A ; 115(25): E5696-E5705, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29866846

RESUMEN

Recent studies have revealed pronounced effects of the spatial distribution of EphA2 receptors on cellular response to receptor activation. However, little is known about molecular mechanisms underlying this spatial sensitivity, in part due to lack of experimental systems. Here, we introduce a hybrid live-cell patterned supported lipid bilayer experimental platform in which the sites of EphA2 activation and integrin adhesion are spatially controlled. Using a series of live-cell imaging and single-molecule tracking experiments, we map the transmission of signals from ephrinA1:EphA2 complexes. Results show that ligand-dependent EphA2 activation induces localized myosin-dependent contractions while simultaneously increasing focal adhesion dynamics throughout the cell. Mechanistically, Src kinase is activated at sites of ephrinA1:EphA2 clustering and subsequently diffuses on the membrane to focal adhesions, where it up-regulates FAK and paxillin tyrosine phosphorylation. EphrinA1:EphA2 signaling triggers multiple cellular responses with differing spatial dependencies to enable a directed migratory response to spatially resolved contact with ephrinA1 ligands.


Asunto(s)
Adhesión Celular/fisiología , Movimiento Celular/fisiología , Efrina-A1/metabolismo , Adhesiones Focales/metabolismo , Adhesiones Focales/fisiología , Receptor EphA2/metabolismo , Transducción de Señal/fisiología , Línea Celular Tumoral , Humanos , Ligandos , Membrana Dobles de Lípidos/metabolismo , Miosinas/metabolismo , Paxillin/metabolismo , Fosforilación/fisiología , Regulación hacia Arriba/fisiología , Familia-src Quinasas/metabolismo
15.
Biophys J ; 114(1): 137-145, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29320680

RESUMEN

Ras is a membrane-anchored signaling protein that serves as a hub for many signaling pathways and also plays a prominent role in cancer. The intrinsic behavior of Ras on the membrane has captivated the biophysics community in recent years, especially the possibility that it may form dimers. In this article, we describe results from a comprehensive series of experiments using fluorescence correlation spectroscopy and single-molecule tracking to probe the possible dimerization of natively expressed and fully processed K-Ras4B in supported lipid bilayer membranes. Key to these studies is the fact that K-Ras4B has its native membrane anchor, including both the farnesylation and methylation of the terminal cysteine, enabling detailed exploration of possible effects of cholesterol and lipid composition on K-Ras4B membrane organization. The results from all conditions studied indicate that full-length K-Ras4B lacks intrinsic dimerization capability. This suggests that any lateral organization of Ras in living cell membranes likely stems from interactions with other factors.


Asunto(s)
Membrana Celular/química , Proteínas Proto-Oncogénicas p21(ras)/química , Humanos , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Propiedades de Superficie
16.
Curr Opin Cell Biol ; 51: 97-102, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29289897

RESUMEN

Receptor tyrosine kinases (RTKs), such as the EGF receptor family, and adhesion molecules, such as integrins, have historically been viewed to have distinctly separable roles in the cell. In this classical view, integrins mediate mechanical interactions between the cell and its surrounding extracellular matrix while RTKs handle signaling to modulate cellular behavior. Although crosstalk between these receptor pathways has been known to exist for a long time, this has generally been attributed to effects significantly downstream from the receptors themselves. In recent years, however, EGFR family RTKs have been found to directly participate in integrin-mediated force sensing, revealing a more complex interplay among these cellular components than originally appreciated. Here we briefly review the classical understanding of EGFR family RTK signaling and then provide a broadened perspective based on recent results.


Asunto(s)
Familia-src Quinasas/metabolismo , Receptores ErbB/metabolismo , Humanos , Transducción de Señal
17.
Biophys J ; 111(5): 1044-52, 2016 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-27602732

RESUMEN

Mechanotransduction at E-cadherin junctions has been postulated to be mediated in part by a force-dependent conformational activation of α-catenin. Activation of α-catenin allows it to interact with vinculin in addition to F-actin, resulting in a strengthening of junctions. Here, using E-cadherin adhesions reconstituted on synthetic, nanopatterned membranes, we show that activation of α-catenin is dependent on E-cadherin clustering, and is sustained in the absence of mechanical force or association with F-actin or vinculin. Adhesions were formed by filopodia-mediated nucleation and micron-scale assembly of E-cadherin clusters, which could be distinguished as either peripheral or central assemblies depending on their relative location at the cell-bilayer adhesion. Whereas F-actin, vinculin, and phosphorylated myosin light chain associated only with the peripheral assemblies, activated α-catenin was present in both peripheral and central assemblies, and persisted in the central assemblies in the absence of actomyosin tension. Impeding filopodia-mediated nucleation and micron-scale assembly of E-cadherin adhesion complexes by confining the movement of bilayer-bound E-cadherin on nanopatterned substrates reduced the levels of activated α-catenin. Taken together, these results indicate that although the initial activation of α-catenin requires micron-scale clustering that may allow the development of mechanical forces, sustained force is not required for maintaining α-catenin in the active state.


Asunto(s)
Cadherinas/metabolismo , Adhesión Celular/fisiología , Mecanotransducción Celular/fisiología , alfa Catenina/metabolismo , Actinas/química , Actinas/metabolismo , Antígenos CD , Cadherinas/química , Cadherinas/genética , Adhesión Celular/efectos de los fármacos , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Membrana Dobles de Lípidos/química , Mecanotransducción Celular/efectos de los fármacos , Microscopía Confocal , Microscopía Fluorescente , Cadenas Ligeras de Miosina/química , Cadenas Ligeras de Miosina/metabolismo , Estrés Mecánico , Vinculina/química , Vinculina/metabolismo , alfa Catenina/química , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/química , Quinasas Asociadas a rho/metabolismo
18.
Nat Struct Mol Biol ; 23(9): 838-46, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27501536

RESUMEN

SOS is a key activator of the small GTPase Ras. In cells, SOS-Ras signaling is thought to be initiated predominantly by membrane recruitment of SOS via the adaptor Grb2 and balanced by rapidly reversible Grb2-SOS binding kinetics. However, SOS has multiple protein and lipid interactions that provide linkage to the membrane. In reconstituted-membrane experiments, these Grb2-independent interactions were sufficient to retain human SOS on the membrane for many minutes, during which a single SOS molecule could processively activate thousands of Ras molecules. These observations raised questions concerning how receptors maintain control of SOS in cells and how membrane-recruited SOS is ultimately released. We addressed these questions in quantitative assays of reconstituted SOS-deficient chicken B-cell signaling systems combined with single-molecule measurements in supported membranes. These studies revealed an essentially one-way trafficking process in which membrane-recruited SOS remains trapped on the membrane and continuously activates Ras until being actively removed via endocytosis.


Asunto(s)
Proteína SOS1/fisiología , Proteínas ras/metabolismo , Regulación Alostérica , Animales , Membrana Celular/metabolismo , Pollos , Endocitosis , Activación Enzimática , Humanos , Células Jurkat , Cinética , Membrana Dobles de Lípidos/química , Sistema de Señalización de MAP Quinasas , Unión Proteica , Dominios Proteicos , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-bcr/fisiología , Proteína SOS1/química , Proteínas ras/química
19.
J Phys Chem B ; 120(5): 867-76, 2016 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-26771210

RESUMEN

The spatial organization of lipid-anchored proteins in the plasma membrane directly influences cell signaling, but measuring such organization in situ is experimentally challenging. The canonical oncogene, c-Src, is a lipid anchored protein that plays a key role in integrin-mediated signal transduction within focal adhesions and cell-cell junctions. Because of its activity in specific plasma membrane regions, structural motifs within the protein have been hypothesized to play an important role in its subcellular localization. This study used a combination of time-resolved fluorescence fluctuation spectroscopy and super-resolution microscopy to quantify the dynamic organization of c-Src in live cell membranes. Pulsed-interleaved excitation fluorescence cross-correlation spectroscopy (PIE-FCCS) showed that a small fraction of c-Src transiently sorts into membrane clusters that are several times larger than the monomers. Photoactivated localization microscopy (PALM) confirmed that c-Src partitions into clusters with low probability and showed that the characteristic size of the clusters is 10-80 nm. Finally, time-resolved fluorescence anisotropy measurements were used to quantify the rotational mobility of c-Src to determine how it interacts with its local environment. Taken together, these results build a quantitative description of the mobility and clustering behavior of the c-Src nonreceptor tyrosine kinase in the live cell plasma membrane.


Asunto(s)
Ácido Mirístico/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Membrana Celular/metabolismo
20.
J Am Chem Soc ; 138(6): 1800-3, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26812279

RESUMEN

Ras, a small GTPase found primarily on the inner leaflet of the plasma membrane, is an important signaling node and an attractive target for anticancer therapies. Lateral organization of Ras on cellular membranes has long been a subject of intense research; in particular, whether it forms dimers on membranes as part of its regulatory function has been a point of great interest. Here we report Ras dimer formation on membranes by Type II photosensitization reactions, in which molecular oxygen mediates the radicalization of proteins under typical fluorescence experimental conditions. The presence of Ras dimers on membranes was detected by diffusion-based fluorescence techniques including fluorescence correlation spectroscopy and single particle tracking, and molecular weights of the stable covalently coupled species were confirmed by gel electrophoresis. Fluorescence spectroscopy implicates interprotein dityrosine as one of the dimerization motifs. The specific surface tyrosine distribution on Ras renders the protein especially sensitive to this reaction, and point mutations affecting surface tyrosines are observed to alter dimerization potential. The photosensitization reactions are reflective of physiological oxidative stress induced by reactive oxygen species, suggesting such processes may occur naturally and influence signaling pathways in cells.


Asunto(s)
Fármacos Fotosensibilizantes/química , Proteínas ras/química , Dimerización , Electroforesis en Gel de Poliacrilamida , Oxidación-Reducción , Espectrometría de Fluorescencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA