Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
NPJ Precis Oncol ; 3: 2, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30675517

RESUMEN

Cancer genomic data is continually growing in complexity, necessitating improved methods for data capture and analysis. Tumors often contain multiple therapeutically relevant alterations, and co-occurring alterations may have a different influence on therapeutic response compared to if those alterations were present alone. One clinically important example of this is the existence of a resistance conferring alteration in combination with a therapeutic sensitizing mutation. The JAX Clinical Knowledgebase (JAX-CKB) (https://ckb.jax.org/) has incorporated the concept of the complex molecular profile, which enables association of therapeutic efficacy data with multiple genomic alterations simultaneously. This provides a mechanism for rapid and accurate assessment of complex cancer-related data, potentially aiding in streamlined clinical decision making. Using the JAX-CKB, we demonstrate the utility of associating data with complex profiles comprising ALK fusions with another variant, which have differing impacts on sensitivity to various ALK inhibitors depending on context.

2.
Mol Biol Cell ; 28(23): 3371-3382, 2017 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-28954861

RESUMEN

The bioactive sphingosine-1-phosphatephosphate (S1P) is present in plasma, bound to carrier proteins, and involved in many physiological processes, including angiogenesis, inflammatory responses, and vascular stabilization. S1P can bind to several G-protein-coupled receptors (GPCRs) activating a number of different signaling networks. At present, the dynamics and relative importance of signaling events activated immediately downstream of GPCR activation are unclear. To examine these, we used a set of fluorescence resonance energy transfer-based biosensors for different RhoGTPases (Rac1, RhoA/B/C, and Cdc42) as well as for heterotrimeric G-proteins in a series of live-cell imaging experiments in primary human endothelial cells. These experiments were accompanied by biochemical GTPase activity assays and transendothelial resistance measurements. We show that S1P promotes cell spreading and endothelial barrier function through S1PR1-Gαi-Rac1 and S1PR1-Gαi-Cdc42 pathways. In parallel, a S1PR2-Gα12/13-RhoA pathway is activated that can induce cell contraction and loss of barrier function, but only if Gαi-mediated signaling is suppressed. Our results suggest that Gαq activity is not involved in S1P-mediated regulation of barrier integrity. Moreover, we show that early activation of RhoA by S1P inactivates Rac1 but not Cdc42, and vice versa. Together, our data show that the rapid S1P-induced increase in endothelial integrity is mediated by a S1PR1-Gαi-Cdc42 pathway.


Asunto(s)
Lisofosfolípidos/metabolismo , Lisofosfolípidos/fisiología , Esfingosina/análogos & derivados , Células Cultivadas , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Células HeLa , Células Endoteliales de la Vena Umbilical Humana , Humanos , Fosforilación , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal/genética , Esfingosina/metabolismo , Esfingosina/fisiología , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
3.
Nat Commun ; 8: 15831, 2017 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-28635959

RESUMEN

During bone remodelling, osteoclasts induce chemotaxis of osteoblasts and yet maintain spatial segregation. We show that osteoclasts express the repulsive guidance factor Semaphorin 4D and induce contact inhibition of locomotion (CIL) in osteoblasts through its receptor Plexin-B1. To examine causality and elucidate how localized Plexin-B1 stimulation may spatiotemporally coordinate its downstream targets in guiding cell migration, we develop an optogenetic tool for Plexin-B1 designated optoPlexin. Precise optoPlexin activation at the leading edge of migrating osteoblasts readily induces local retraction and, unexpectedly, distal protrusions to steer cells away. These morphological changes are accompanied by reorganization of Myosin II, PIP3, adhesion and active Cdc42. We attribute the resultant repolarization to RhoA/ROCK-mediated redistribution of ß-Pix, which activates Cdc42 and promotes protrusion. Thus, our data demonstrate a causal role of Plexin-B1 for CIL in osteoblasts and reveals a previously unknown effect of Semaphorin signalling on spatial distribution of an activator of cell migration.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Movimiento Celular/efectos de la radiación , Polaridad Celular/efectos de la radiación , Luz , Masculino , Ratones , Ratones Endogámicos C57BL , Miosina Tipo II/genética , Miosina Tipo II/metabolismo , Proteínas del Tejido Nervioso/genética , Optogenética , Osteoblastos/citología , Osteoblastos/efectos de la radiación , Osteoclastos/citología , Osteoclastos/efectos de la radiación , Receptores de Superficie Celular/genética , Semaforinas/metabolismo , Transducción de Señal/efectos de la radiación , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
4.
Mol Biol Cell ; 28(10): 1347-1360, 2017 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-28356423

RESUMEN

Breast carcinoma cells use specialized, actin-rich protrusions called invadopodia to degrade and invade through the extracellular matrix. Phosphorylation of the actin nucleation-promoting factor and actin-stabilizing protein cortactin downstream of the epidermal growth factor receptor-Src-Arg kinase cascade is known to be a critical trigger for invadopodium maturation and subsequent cell invasion in breast cancer cells. The functions of cortactin phosphorylation in this process, however, are not completely understood. We identify the Rho-family guanine nucleotide exchange factor Vav2 in a comprehensive screen for human SH2 domains that bind selectively to phosphorylated cortactin. We demonstrate that the Vav2 SH2 domain binds selectively to phosphotyrosine-containing peptides corresponding to cortactin tyrosines Y421 and Y466 but not to Y482. Mutation of the Vav2 SH2 domain disrupts its recruitment to invadopodia, and an SH2-domain mutant form of Vav2 cannot support efficient matrix degradation in invasive MDA-MB-231 breast cancer cells. We show that Vav2 function is required for promoting invadopodium maturation and consequent actin polymerization, matrix degradation, and invasive migratory behavior. Using biochemical assays and a novel Rac3 biosensor, we show that Vav2 promotes Rac3 activation at invadopodia. Rac3 knockdown reduces matrix degradation by invadopodia, whereas a constitutively active Rac3 can rescue the deficits in invadopodium function in Vav2-knockdown cells. Together these data indicate that phosphorylated cortactin recruits Vav2 to activate Rac3 and promote invadopodial maturation in invasive breast cancer cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Cortactina/metabolismo , Podosomas/metabolismo , Proteínas Proto-Oncogénicas c-vav/metabolismo , Actinas/metabolismo , Línea Celular Tumoral , Matriz Extracelular/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Invasividad Neoplásica , Fosforilación , Fosfotirosina/metabolismo , Podosomas/fisiología , Proteínas Tirosina Quinasas/metabolismo , Seudópodos/metabolismo , Proteínas de Unión al GTP rac/metabolismo
5.
Sci Rep ; 6: 36825, 2016 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-27833100

RESUMEN

Rho GTPases are master regulators of the eukaryotic cytoskeleton. The activation of Rho GTPases is governed by Rho guanine nucleotide exchange factors (GEFs). Three RhoGEF isoforms are produced by the gene ARHGEF25; p63RhoGEF580, GEFT and a recently discovered longer isoform of 619 amino acids (p63RhoGEF619). The subcellular distribution of p63RhoGEF580 and p63RhoGEF619 is strikingly different in unstimulated cells, p63RhoGEF580 is located at the plasma membrane and p63RhoGEF619 is confined to the cytoplasm. Interestingly, we find that both P63RhoGEF580 and p63RhoGEF619 activate RhoGTPases to a similar extent after stimulation of Gαq coupled GPCRs. Furthermore, we show that p63RhoGEF619 relocates to the plasma membrane upon activation of Gαq coupled GPCRs, resembling the well-known activation mechanism of RhoGEFs activated by Gα12/13. Synthetic recruitment of p63RhoGEF619 to the plasma membrane increases RhoGEF activity towards RhoA, but full activation requires allosteric activation via Gαq. Together, these findings reveal a dual role for Gαq in RhoGEF activation, as it both recruits and allosterically activates cytosolic ARHGEF25 isoforms.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/fisiología , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Regulación Alostérica , Secuencia de Aminoácidos , Membrana Celular/metabolismo , Células HeLa , Humanos , Cinética , Isoformas de Proteínas/metabolismo , Transporte de Proteínas , Transducción de Señal
6.
Sci Rep ; 6: 25502, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-27147504

RESUMEN

Endothelial cells line the vasculature and are important for the regulation of blood pressure, vascular permeability, clotting and transendothelial migration of leukocytes and tumor cells. A group of proteins that that control the endothelial barrier function are the RhoGTPases. This study focuses on three homologous (>88%) RhoGTPases: RhoA, RhoB, RhoC of which RhoB and RhoC have been poorly characterized. Using a RhoGTPase mRNA expression analysis we identified RhoC as the highest expressed in primary human endothelial cells. Based on an existing RhoA FRET sensor we developed new RhoB/C FRET sensors to characterize their spatiotemporal activation properties. We found all these RhoGTPase sensors to respond to physiologically relevant agonists (e.g. Thrombin), reaching transient, localized FRET ratio changes up to 200%. These RhoA/B/C FRET sensors show localized GEF and GAP activity and reveal spatial activation differences between RhoA/C and RhoB. Finally, we used these sensors to monitor GEF-specific differential activation of RhoA/B/C. In summary, this study adds high-contrast RhoB/C FRET sensors to the currently available FRET sensor toolkit and uncover new insights in endothelial and RhoGTPase cell biology. This allows us to study activation and signaling by these closely related RhoGTPases with high spatiotemporal resolution in primary human cells.


Asunto(s)
Antígenos CD/genética , Técnicas Biosensibles/métodos , Cadherinas/genética , Células Endoteliales de la Vena Umbilical Humana/enzimología , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoB/genética , Proteína rhoC de Unión a GTP/genética , Antígenos CD/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cadherinas/metabolismo , Activación Enzimática , Transferencia Resonante de Energía de Fluorescencia , Uniones Comunicantes , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Modelos Moleculares , Nocodazol/farmacología , Cultivo Primario de Células , Estructura Secundaria de Proteína , Transducción de Señal , Trombina/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Proteína de Unión al GTP rhoA/metabolismo , Proteína de Unión al GTP rhoB/metabolismo , Proteína rhoC de Unión a GTP/metabolismo
7.
Nat Commun ; 7: 10493, 2016 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-26814335

RESUMEN

During immune surveillance and inflammation, leukocytes exit the vasculature through transient openings in the endothelium without causing plasma leakage. However, the exact mechanisms behind this intriguing phenomenon are still unknown. Here we report that maintenance of endothelial barrier integrity during leukocyte diapedesis requires local endothelial RhoA cycling. Endothelial RhoA depletion in vitro or Rho inhibition in vivo provokes neutrophil-induced vascular leakage that manifests during the physical movement of neutrophils through the endothelial layer. Local RhoA activation initiates the formation of contractile F-actin structures that surround emigrating neutrophils. These structures that surround neutrophil-induced endothelial pores prevent plasma leakage through actomyosin-based pore confinement. Mechanistically, we found that the initiation of RhoA activity involves ICAM-1 and the Rho GEFs Ect2 and LARG. In addition, regulation of actomyosin-based endothelial pore confinement involves ROCK2b, but not ROCK1. Thus, endothelial cells assemble RhoA-controlled contractile F-actin structures around endothelial pores that prevent vascular leakage during leukocyte extravasation.


Asunto(s)
Actinas/metabolismo , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Leucocitos/citología , Transducción de Señal , Migración Transendotelial y Transepitelial , Proteína de Unión al GTP rhoA/metabolismo , Citoesqueleto de Actina/química , Citoesqueleto de Actina/metabolismo , Actinas/genética , Animales , Permeabilidad Capilar , Células Cultivadas , Endotelio Vascular/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular , Leucocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Neutrófilos/citología , Neutrófilos/metabolismo , Proteína de Unión al GTP rhoA/genética
8.
J Biol Chem ; 291(9): 4323-33, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26740622

RESUMEN

Invadosomes are actin-rich membrane protrusions that degrade the extracellular matrix to drive tumor cell invasion. Key players in invadosome formation are c-Src and Rho family GTPases. Invadosomes can reassemble into circular rosette-like superstructures, but the underlying signaling mechanisms remain obscure. Here we show that Src-induced invadosomes in human melanoma cells (A375M and MDA-MB-435) undergo rapid remodeling into dynamic extracellular matrix-degrading rosettes by distinct G protein-coupled receptor agonists, notably lysophosphatidic acid (LPA; acting through the LPA1 receptor) and endothelin. Agonist-induced rosette formation is blocked by pertussis toxin, dependent on PI3K activity and accompanied by localized production of phosphatidylinositol 3,4,5-trisphosphate, whereas MAPK and Ca(2+) signaling are dispensable. Using FRET-based biosensors, we show that LPA and endothelin transiently activate Cdc42 through Gi, concurrent with a biphasic decrease in Rac activity and differential effects on RhoA. Cdc42 activity is essential for rosette formation, whereas G12/13-mediated RhoA-ROCK signaling suppresses the remodeling process. Our results reveal a Gi-mediated Cdc42 signaling axis by which G protein-coupled receptors trigger invadosome remodeling, the degree of which is dictated by the Cdc42-RhoA activity balance.


Asunto(s)
Endotelinas/metabolismo , Lisofosfolípidos/metabolismo , Melanoma/metabolismo , Podosomas/metabolismo , Receptores del Ácido Lisofosfatídico/agonistas , Proteína de Unión al GTP cdc42/agonistas , Proteína de Unión al GTP rac1/metabolismo , Biomarcadores/metabolismo , Línea Celular Tumoral , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Transferencia Resonante de Energía de Fluorescencia , Humanos , Hidrólisis , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Melanoma/enzimología , Melanoma/patología , Microscopía Confocal , Microscopía Fluorescente , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Podosomas/enzimología , Podosomas/patología , Interferencia de ARN , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores del Ácido Lisofosfatídico/antagonistas & inhibidores , Receptores del Ácido Lisofosfatídico/genética , Receptores del Ácido Lisofosfatídico/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Imagen de Lapso de Tiempo , Proteína de Unión al GTP cdc42/antagonistas & inhibidores , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/agonistas , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
9.
Sci Rep ; 5: 14693, 2015 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-26435194

RESUMEN

The small GTPase RhoA is involved in cell morphology and migration. RhoA activity is tightly regulated in time and space and depends on guanine exchange factors (GEFs). However, the kinetics and subcellular localization of GEF activity towards RhoA are poorly defined. To study the mechanism underlying the spatiotemporal control of RhoA activity by GEFs, we performed single cell imaging with an improved FRET sensor reporting on the nucleotide loading state of RhoA. By employing the FRET sensor we show that a plasma membrane located RhoGEF, p63RhoGEF, can rapidly activate RhoA through endogenous GPCRs and that localized RhoA activity at the cell periphery correlates with actin polymerization. Moreover, synthetic recruitment of the catalytic domain derived from p63RhoGEF to the plasma membrane, but not to the Golgi apparatus, is sufficient to activate RhoA. The synthetic system enables local activation of endogenous RhoA and effectively induces actin polymerization and changes in cellular morphology. Together, our data demonstrate that GEF activity at the plasma membrane is sufficient for actin polymerization via local RhoA signaling.


Asunto(s)
Actinas/metabolismo , Membrana Celular/enzimología , Proteína de Unión al GTP rhoA/fisiología , Núcleo Celular , Activación Enzimática , Células HeLa , Humanos , Multimerización de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Factores de Intercambio de Guanina Nucleótido Rho , Factores de Transcripción/metabolismo
11.
J Cell Sci ; 128(16): 3041-54, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26116572

RESUMEN

Endothelial cell-cell junctions maintain a restrictive barrier that is tightly regulated to allow dynamic responses to permeability-inducing angiogenic factors, as well as to inflammatory agents and adherent leukocytes. The ability of these stimuli to transiently remodel adherens junctions depends on Rho-GTPase-controlled cytoskeletal rearrangements. How the activity of Rho-GTPases is spatio-temporally controlled at endothelial adherens junctions by guanine-nucleotide exchange factors (GEFs) is incompletely understood. Here, we identify a crucial role for the Rho-GEF Trio in stabilizing junctions based around vascular endothelial (VE)-cadherin (also known as CDH5). Trio interacts with VE-cadherin and locally activates Rac1 at adherens junctions during the formation of nascent contacts, as assessed using a novel FRET-based Rac1 biosensor and biochemical assays. The Rac-GEF domain of Trio is responsible for the remodeling of junctional actin from radial into cortical actin bundles, a crucial step for junction stabilization. This promotes the formation of linear adherens junctions and increases endothelial monolayer resistance. Collectively, our data show the importance of spatio-temporal regulation of the actin cytoskeleton through Trio and Rac1 at VE-cadherin-based cell-cell junctions in the maintenance of the endothelial barrier.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Uniones Intercelulares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Antígenos CD/genética , Cadherinas/genética , Permeabilidad Capilar/genética , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , GTP Fosfohidrolasas/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Uniones Intercelulares/genética , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal/genética , Proteína de Unión al GTP rac1/genética
12.
Nat Commun ; 6: 6619, 2015 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-25851023

RESUMEN

Directed cell migration in native environments is influenced by multiple migratory cues. These cues may include simultaneously occurring attractive soluble growth factor gradients and repulsive effects arising from cell-cell contact, termed contact inhibition of locomotion (CIL). How single cells reconcile potentially conflicting cues remains poorly understood. Here we show that a dynamic crosstalk between epidermal growth factor (EGF)-mediated chemotaxis and CIL guides metastatic breast cancer cell motility, whereby cells become progressively insensitive to CIL in a chemotactic input-dependent manner. This balance is determined via integration of protrusion-enhancing signalling from EGF gradients and protrusion-suppressing signalling induced by CIL, mediated in part through EphB. Our results further suggest that EphB and EGF signalling inputs control protrusion formation by converging onto regulation of phosphatidylinositol 3-kinase (PI3K). We propose that this intricate interplay may enhance the spread of loose cell ensembles in pathophysiological conditions such as cancer, and possibly other physiological settings.


Asunto(s)
Movimiento Celular/genética , Quimiotaxis/genética , Inhibición de Contacto/fisiología , Neoplasias Mamarias Animales/genética , Receptor EphB3/genética , Animales , Western Blotting , Línea Celular Tumoral , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Neoplasias Mamarias Animales/metabolismo , Ratas , Receptores de la Familia Eph/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Proc Natl Acad Sci U S A ; 112(3): E267-76, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25561548

RESUMEN

Polarized Rac1 signaling is a hallmark of many cellular functions, including cell adhesion, motility, and cell division. The two steps of Rac1 activation are its translocation to the plasma membrane and the exchange of nucleotide from GDP to GTP. It is, however, unclear whether these two processes are regulated independent of each other and what their respective roles are in polarization of Rac1 signaling. We designed a single-particle tracking (SPT) method to quantitatively analyze the kinetics of Rac1 membrane translocation in living cells. We found that the rate of Rac1 translocation was significantly elevated in protrusions during cell spreading on collagen. Furthermore, combining FRET sensor imaging with SPT measurements in the same cell, the recruitment of Rac1 was found to be polarized to an extent similar to that of the nucleotide exchange process. Statistical analysis of single-molecule trajectories and optogenetic manipulation of membrane lipids revealed that Rac1 membrane translocation precedes nucleotide exchange, and is governed primarily by interactions with phospholipids, particularly PI(3,4,5)P3, instead of protein factors. Overall, the study highlights the significance of membrane translocation in spatial Rac1 signaling, which is in addition to the traditional view focusing primarily on GEF distribution and exchange reaction.


Asunto(s)
Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo , Humanos , Cinética , Células MCF-7 , Microscopía Fluorescente , Transporte de Proteínas
14.
Methods Mol Biol ; 1251: 277-89, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25391805

RESUMEN

Recent developments in optogenetics have extended optical control of signaling to intracellular proteins, including Rac, a small G protein in the Rho family. A blue light-sensing LOV (light, oxygen, or voltage) domain derived from Avena sativa (oat) phototropin was fused to the N-terminus of a constitutively active mutant of Rac, via an α-helix (Jα) that is conserved among plant phototropins. The fused LOV domain occluded binding of downstream effectors to Rac in the dark. Exposure to blue light caused a conformational change of the LOV domain and unwinding of the Jα helix, relieving steric inhibition. The LOV domain incorporates a flavin as the photon-absorbing cofactor and can be activated by light in a reversible and repeatable fashion. In cultured cells, global illumination with blue light rapidly activated Rac and led to cell spreading and membrane ruffling. Localized and pulsed illumination generated a gradient of Rac activity and induced directional migration. In this chapter, we will describe the techniques in detail and present some examples of applications of using photoactivatable Rac (PA-Rac) in living cells.


Asunto(s)
Técnicas Citológicas/métodos , Microscopía Fluorescente/métodos , Optogenética/métodos , Transducción de Señal/fisiología , Proteínas de Unión al GTP rac/ultraestructura , Animales , Avena/química , Células HeLa , Humanos , Luz , Iluminación , Ratones , Microscopía Fluorescente/instrumentación , Fototropinas/análisis , Fototropinas/genética , Conformación Proteica , Estructura Terciaria de Proteína
15.
Pflugers Arch ; 465(3): 397-408, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23417571

RESUMEN

Optogenetics arises from the innovative application of microbial opsins in mammalian neurons and has since been a powerful technology that fuels the advance of our knowledge in neuroscience. In recent years, there has been growing interest in designing optogenetic tools extendable to broader cell types and biochemical signals. To date, a variety of photoactivatable proteins (refers to induction of protein activity in contrast to fluorescence) have been developed based on the understanding of plant and microbial photoreceptors including phototropins, blue light sensors using flavin adenine dinucleotide proteins, cryptochromes, and phytochromes. Such tools offered researchers reversible, quantitative, and precise spatiotemporal control of enzymatic activity, protein-protein interaction, protein translocation, as well as gene transcription in cells and in whole animals. In this review, we will briefly introduce these photosensory proteins, describe recent developments in optogenetics, and compare and contrast different methods based on their advantages and limitations.


Asunto(s)
Optogenética , Transducción de Señal , Regulación Alostérica , Animales , Bacterias , Criptocromos/genética , Criptocromos/metabolismo , Humanos , Fotorreceptores Microbianos/genética , Fotorreceptores Microbianos/metabolismo , Fototropinas/genética , Fototropinas/metabolismo , Plantas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
16.
J Cell Sci ; 123(Pt 20): 3576-86, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20876660

RESUMEN

We previously showed that the cell-cell junction protein plakoglobin (PG) not only suppresses motility of keratinocytes in contact with each other, but also, unexpectedly, of single cells. Here we show that PG deficiency results in extracellular matrix (ECM)-dependent disruption of mature focal adhesions and cortical actin organization. Plating PG⁻/⁻ cells onto ECM deposited by PG+/⁻ cells partially restored normal cell morphology and inhibited PG⁻/⁻ cell motility. In over 70 adhesion molecules whose expression we previously showed to be altered in PG⁻/⁻ cells, a substantial decrease in fibronectin (FN) in PG⁻/⁻ cells stood out. Re-introduction of PG into PG⁻/⁻ cells restored FN expression, and keratinocyte motility was reversed by plating PG⁻/⁻ cells onto FN. Somewhat surprisingly, based on previously reported roles for PG in regulating gene transcription, PG-null cells exhibited an increase, not a decrease, in FN promoter activity. Instead, PG was required for maintenance of FN mRNA stability. PG⁻/⁻ cells exhibited an increase in activated Src, one of the kinases controlled by FN, a phenotype reversed by plating PG⁻/⁻ cells on ECM deposited by PG+/⁻ keratinocytes. PG⁻/⁻ cells also exhibited Src-independent activation of the small GTPases Rac1 and RhoA. Both Src and RhoA inhibition attenuated PG⁻/⁻ keratinocyte motility. We propose a novel role for PG in regulating cell motility through distinct ECM-Src and RhoGTPase-dependent pathways, influenced in part by PG-dependent regulation of FN mRNA stability.


Asunto(s)
Movimiento Celular/fisiología , Fibronectinas/metabolismo , Transducción de Señal/fisiología , gamma Catenina/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Western Blotting , Movimiento Celular/genética , Células Cultivadas , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Fibronectinas/genética , Técnica del Anticuerpo Fluorescente Indirecta , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , gamma Catenina/genética , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/genética
17.
Mol Cell Proteomics ; 9(2): 351-61, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19955077

RESUMEN

The ability of cells to modulate interactions with each other and the substrate is essential for epithelial tissue remodeling during processes such as wound healing and tumor progression. However, despite strides made in the field of proteomics, proteins involved in adhesion have been difficult to study. Here, we report a method for the enrichment and analysis of proteins associated with the basal surface of the cell and its underlying matrix. The enrichment involves deroofing the cells with 20 mM ammonium hydroxide and the removal of cytosolic and organellar proteins by stringent water wash. Proteomic profiling was achieved by LC-FTMS, which allowed comparison of differentially expressed or shared proteins under different cell states. First, we analyzed and compared the basal cell components of mouse keratinocytes lacking the cell-cell junction molecule plakoglobin with their control counterparts. Changes in the molecules involved in motility and invasion were detected in plakoglobin-deficient cells, including decreased detection of fibronectin, integrin beta(4), and FAT tumor suppressor. Second, we assessed the differences in basal cell components between two human oral squamous cell carcinoma lines originating from different sites in the oral cavity (CAL33 and UM-SCC-1). The data show differences between the two lines in the type and abundance of proteins specific to cell adhesion, migration, and angiogenesis. Therefore, the method described here has the potential to serve as a platform to assess proteomic changes in basal cell components including extracellular and adhesion-specific proteins involved in wound healing, cancer, and chronic and acquired adhesion-related disorders.


Asunto(s)
Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Queratinocitos/metabolismo , Espectrometría de Masas/métodos , Proteínas/genética , Proteínas/metabolismo , Hidróxido de Amonio , Animales , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Humanos , Hidróxidos/farmacología , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Ratones , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Péptidos/química , Péptidos/metabolismo , gamma Catenina/deficiencia , gamma Catenina/metabolismo
18.
Curr Biol ; 19(11): 967-73, 2009 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-19398338

RESUMEN

Although many proteins, receptors, and viruses are transported rearward along filopodia by retrograde actin flow, it is less clear how molecules move forward in filopodia. Myosin-X (Myo10) is an actin-based motor hypothesized to use its motor activity to move forward along actin filaments to the tips of filopodia. Here we use a sensitive total internal reflection fluorescence (TIRF) microscopy system to directly visualize the movements of GFP-Myo10. This reveals a novel form of motility at or near the single-molecule level in living cells wherein extremely faint particles of Myo10 move in a rapid and directed fashion toward the filopodial tip. These fast forward movements occur at approximately 600 nm/s over distances of up to approximately 10 microm and require Myo10 motor activity and actin filaments. As expected for imaging at the single-molecule level, the faint particles of GFP-Myo10 are diffraction limited, have an intensity range similar to single GFP molecules, and exhibit stepwise bleaching. Faint particles of GFP-Myo5a can also move toward the filopodial tip, but at a slower characteristic velocity of approximately 250 nm/s. Similar movements were not detected with GFP-Myo1a, indicating that not all myosins are capable of intrafilopodial motility. These data indicate the existence of a novel system of long-range transport based on the rapid movement of myosin molecules along filopodial actin filaments.


Asunto(s)
Movimiento Celular/fisiología , Miosinas/fisiología , Seudópodos/fisiología , Actinas/fisiología , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Bovinos , Movimiento Celular/efectos de los fármacos , Proteínas Fluorescentes Verdes/análisis , Células HeLa , Humanos , Microscopía Fluorescente , Miosinas/análisis , Miosinas/ultraestructura , Seudópodos/metabolismo , Seudópodos/ultraestructura , Tiazolidinas/farmacología
19.
Cell Commun Adhes ; 15(4): 333-49, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18979298

RESUMEN

Endothelial cells (ECs) express VE-cadherin and N-cadherin, and recent data suggest that VE-cadherin levels are dependent on N-cadherin expression. While investigating changes in N-cadherin levels during endothelial monolayer maturation, the authors found that VE-cadherin levels are maintained in ECs despite a decrease in N-cadherin, suggesting that VE-cadherin levels may not depend on N-cadherin. Knockdown of N-cadherin did not affect VE-cadherin levels in ECs with low endogenous N-cadherin expression. Surprisingly, however, knockdown of N-cadherin in ECs with high endogenous N-cadherin expression increased VE-cadherin levels, suggesting an inverse relationship between the two. This was further supported by a decrease in VE-cadherin following overexpression of N-cadherin. Experiments in which p120, a catenin that binds N- and VE-cadherin, was knocked down or overexpressed indicate that these two cadherins compete for p120. These data demonstrate that VE-cadherin levels are not directly related to N-cadherin levels but may be inversely related due to competition for p120.


Asunto(s)
Cadherinas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Endoteliales/metabolismo , Fosfoproteínas/metabolismo , Animales , Antígenos CD/biosíntesis , Antígenos CD/metabolismo , Cadherinas/biosíntesis , Cateninas , Bovinos , Células Cultivadas , Técnicas de Silenciamiento del Gen , Humanos , Interferencia de ARN , ARN Interferente Pequeño , Catenina delta
20.
J Biol Chem ; 280(48): 40355-63, 2005 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-16183992

RESUMEN

Plakoglobin (PG) is a member of the Armadillo family of adhesion/signaling proteins that can be incorporated into both adherens junctions and desmosomes. Loss of PG results in defects in the mechanical integrity of heart and skin and decreased adhesive strength in keratinocyte cultures established from the skin of PG knock-out (PG-/-) mice, the latter of which cannot be compensated for by overexpressing the closely related beta-catenin. In this study, we examined the mechanisms of PG-regulated adhesion in murine keratinocytes. Biochemical and morphological analyses indicated that junctional incorporation of desmosomal, but not adherens junction, components was impaired in PG-/- cells compared with PG+/- controls. Re-expression of PG, but not beta-catenin, in PG-/- cells largely reversed these effects, indicating a key role for PG in desmosome assembly. Epidermal growth factor (EGF) receptor activation resulted in Tyr phosphorylation of PG, which was accompanied by a loss of desmoplakin from desmosomes and decreased adhesive strength following 18-h EGF treatment. Importantly, introduction of a phosphorylation-deficient PG mutant into PG null cells prevented the EGF receptor-dependent loss of desmoplakin from junctions, attenuating the effects of long term EGF treatment on cell adhesion. Therefore, PG is essential for maintaining and regulating adhesive strength in keratinocytes largely through its contributions to desmosome assembly and structure. As a target for modulation by EGF, regulation of PG-dependent adhesion may play an important role during wound healing and tumor metastasis.


Asunto(s)
Desmosomas/metabolismo , Receptores ErbB/metabolismo , gamma Catenina/genética , Adenoviridae/metabolismo , Uniones Adherentes , Animales , Western Blotting , Adhesión Celular , Células Cultivadas , Detergentes/farmacología , Relación Dosis-Respuesta a Droga , Factor de Crecimiento Epidérmico/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Inmunoprecipitación , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Ratones Noqueados , Microscopía Fluorescente , Metástasis de la Neoplasia , Octoxinol/farmacología , Fosforilación , Estructura Terciaria de Proteína , Transducción de Señal , Factores de Tiempo , Tirosina/química , Cicatrización de Heridas , beta Catenina/metabolismo , gamma Catenina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...