Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Cell Dev Biol ; 33: 103-125, 2017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28793794

RESUMEN

Although directed migration of eukaryotic cells may have evolved to escape nutrient depletion, it has been adopted for an extensive range of physiological events during development and in the adult organism. The subversion of these movements results in disease, such as cancer. Mechanisms of propulsion and sensing are extremely diverse, but most eukaryotic cells move by extending actin-filled protrusions termed macropinosomes, pseudopodia, or lamellipodia or by extension of blebs. In addition to motility, directed migration involves polarity and directional sensing. The hundreds of gene products involved in these processes are organized into networks of parallel and interconnected pathways. Many of these components are activated or inhibited coordinately with stimulation and on each spontaneously extended protrusion. Moreover, these networks display hallmarks of excitability, including all-or-nothing responsiveness and wave propagation. Cellular protrusions result from signal transduction waves that propagate outwardly from an origin and drive cytoskeletal activity. The range of the propagating waves and hence the size of the protrusions can be altered by lowering or raising the threshold for network activation, with larger and wider protrusions favoring gliding or oscillatory behavior over amoeboid migration. Here, we evaluate the variety of models of excitable networks controlling directed migration and outline critical tests. We also discuss the utility of this emerging view in producing cell migration and in integrating the various extrinsic cues that direct migration.


Asunto(s)
Movimiento Celular , Transducción de Señal , Animales , Humanos , Modelos Biológicos
2.
Proc Natl Acad Sci U S A ; 120(19): e2218906120, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37126708

RESUMEN

Cellular sensing of most environmental cues involves receptors that affect a signal-transduction excitable network (STEN), which is coupled to a cytoskeletal excitable network (CEN). We show that the mechanism of sensing of nanoridges is fundamentally different. CEN activity occurs preferentially on nanoridges, whereas STEN activity is constrained between nanoridges. In the absence of STEN, waves disappear, but long-lasting F-actin puncta persist along the ridges. When CEN is suppressed, wave propagation is no longer constrained by nanoridges. A computational model reproduces these experimental observations. Our findings indicate that nanotopography is sensed directly by CEN, whereas STEN is only indirectly affected due to a CEN-STEN feedback loop. These results explain why texture sensing is robust and acts cooperatively with multiple other guidance cues in complex, in vivo microenvironments.


Asunto(s)
Citoesqueleto de Actina , Citoesqueleto , Movimiento Celular , Actinas , Microtúbulos
3.
EMBO J ; 40(4): e105094, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33586225

RESUMEN

The ability of cells to polarize and move toward external stimuli plays a crucial role in development, as well as in normal and pathological physiology. Migrating cells maintain dynamic complementary distributions of Ras activity and of the phospholipid phosphatidylinositol-3,4-bisphosphate (PI(3,4)P2). Here, we show that lagging-edge component PI(3,4)P2 also localizes to retracting leading-edge protrusions and nascent macropinosomes, even in the absence of phosphatidylinositol 3,4,5-trisphosphate (PIP3). Once internalized, macropinosomes break up into smaller PI(3,4)P2-enriched vesicles, which fuse with the plasma membrane at the rear of the cell. Subsequently, the phosphoinositide diffuses toward the front of the cell, where it is degraded. Computational modeling confirms that this cycle gives rise to stable back-to-front gradient. These results uncover a surprising "reverse-fountain flow" of PI(3,4)P2 that regulates polarity.


Asunto(s)
Membrana Celular/metabolismo , Movimiento Celular , Dictyostelium/fisiología , Microtúbulos/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Dictyostelium/citología , Células HL-60 , Humanos
4.
Proc Natl Acad Sci U S A ; 117(8): 4158-4168, 2020 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-32051246

RESUMEN

Cancer cells display novel characteristics which can be exploited for therapeutic advantage. Isolated studies have shown that 1) the mevalonate pathway and 2) increased macropinocytosis are important in tumorigenesis, but a connection between these two observations has not been envisioned. A library screen for compounds that selectively killed Dictyostelium pten- cells identified pitavastatin. Pitavastatin also killed human breast epithelial MCF10A cells lacking PTEN or expressing K-RasG12V, as well as mouse tumor organoids. The selective killing of cells with oncogenic defects was traced to GGPP (geranylgeranyl diphosphate) depletion. Disruption of GGPP synthase in Dictyostelium revealed that GGPP is needed for pseudopod extension and macropinocytosis. Fluid-phase uptake through macropinocytosis is lower in PTEN-deleted cells and, as reported previously, higher in cells expressing activated Ras. Nevertheless, uptake was more sensitive to pitavastatin in cells with either of these oncogenic mutations than in wild-type cells. Loading the residual macropinosomes after pitavastatin with high concentrations of protein mitigated the cell death, indicating that defective macropinocytosis leads to amino acid starvation. Our studies suggest that the dependence of cancer cells on the mevalonate pathway is due to the role of GGPP in macropinocytosis and the reliance of these cells on macropinocytosis for nutrient uptake. Thus, inhibition of the networks mediating these processes is likely to be effective in cancer intervention.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Ácido Mevalónico/farmacología , Pinocitosis/efectos de los fármacos , Quinolinas/farmacología , Animales , Línea Celular , Dictyostelium/efectos de los fármacos , Dictyostelium/fisiología , Humanos , Ratones , Oncogenes , Organoides
5.
Semin Cell Dev Biol ; 100: 133-142, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31836289

RESUMEN

The directed movements of individual, groups, or sheets of cells at specific times in particular locations bring about form and complexity to developing organisms. Cells move by extending protrusions, such as macropinosomes, pseudopods, lamellipods, filopods, or blebs. Although many of the cytoskeletal components within these structures are known, less is known about the mechanisms that determine their location, number, and characteristics. Recent evidence suggests that control may be exerted by a signal transduction excitable network whose components and activities, including Ras, PI3K, TorC2, and phosphoinositides, self-organize on the plasma membrane and propagate in waves. The waves drive the various types of protrusions, which in turn, determine the modes of cell migration. Acute perturbations at specific points in the network produce abrupt shifts in protrusion type, including transitions from pseudopods to filopods or lamellipods. These observations have also contributed to a delineation of the signal transduction network, including candidate fast positive and delayed negative feedback loops. The network contains many oncogenes and tumor suppressors, and other molecules which have recently been implicated in developmental and metabolic abnormalities. Thus, the concept of signal transduction network excitability in cell migration can be used to understand disease states and morphological changes occurring in development.


Asunto(s)
Movimiento Celular , Enfermedad , Redes y Vías Metabólicas , Actinas/metabolismo , Animales , Citoesqueleto/metabolismo , Humanos , Transducción de Señal
6.
Mol Syst Biol ; 17(12): e10505, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34898015

RESUMEN

Motile cells can use and switch between different modes of migration. Here, we use traction force microscopy and fluorescent labeling of actin and myosin to quantify and correlate traction force patterns and cytoskeletal distributions in Dictyostelium discoideum cells that move and switch between keratocyte-like fan-shaped, oscillatory, and amoeboid modes. We find that the wave dynamics of the cytoskeletal components critically determine the traction force pattern, cell morphology, and migration mode. Furthermore, we find that fan-shaped cells can exhibit two different propulsion mechanisms, each with a distinct traction force pattern. Finally, the traction force patterns can be recapitulated using a computational model, which uses the experimentally determined spatiotemporal distributions of actin and myosin forces and a viscous cytoskeletal network. Our results suggest that cell motion can be generated by friction between the flow of this network and the substrate.


Asunto(s)
Actomiosina , Dictyostelium , Citoesqueleto de Actina , Actinas , Movimiento Celular , Tracción
7.
PLoS Comput Biol ; 17(7): e1008803, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34260581

RESUMEN

During the last decade, a consensus has emerged that the stochastic triggering of an excitable system drives pseudopod formation and subsequent migration of amoeboid cells. The presence of chemoattractant stimuli alters the threshold for triggering this activity and can bias the direction of migration. Though noise plays an important role in these behaviors, mathematical models have typically ignored its origin and merely introduced it as an external signal into a series of reaction-diffusion equations. Here we consider a more realistic description based on a reaction-diffusion master equation formalism to implement these networks. In this scheme, noise arises naturally from a stochastic description of the various reaction and diffusion terms. Working on a three-dimensional geometry in which separate compartments are divided into a tetrahedral mesh, we implement a modular description of the system, consisting of G-protein coupled receptor signaling (GPCR), a local excitation-global inhibition mechanism (LEGI), and signal transduction excitable network (STEN). Our models implement detailed biochemical descriptions whenever this information is available, such as in the GPCR and G-protein interactions. In contrast, where the biochemical entities are less certain, such as the LEGI mechanism, we consider various possible schemes and highlight the differences between them. Our simulations show that even when the LEGI mechanism displays perfect adaptation in terms of the mean level of proteins, the variance shows a dose-dependence. This differs between the various models considered, suggesting a possible means for determining experimentally among the various potential networks. Overall, our simulations recreate temporal and spatial patterns observed experimentally in both wild-type and perturbed cells, providing further evidence for the excitable system paradigm. Moreover, because of the overall importance and ubiquity of the modules we consider, including GPCR signaling and adaptation, our results will be of interest beyond the field of directed migration.


Asunto(s)
Factores Quimiotácticos/farmacología , Quimiotaxis/efectos de los fármacos , Simulación por Computador , Modelos Biológicos , Biología Computacional , Difusión , Seudópodos/efectos de los fármacos , Procesos Estocásticos
8.
Proc Natl Acad Sci U S A ; 115(16): E3722-E3730, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29602807

RESUMEN

Cell migration requires the coordination of an excitable signal transduction network involving Ras and PI3K pathways with cytoskeletal activity. We show that expressing activated Ras GTPase-family proteins in cells lacking PTEN or other mutations which increase cellular protrusiveness transforms cells into a persistently activated state. Leading- and trailing-edge markers were found exclusively at the cell perimeter and the cytosol, respectively, of the dramatically flattened cells. In addition, the lifetimes of dynamic actin puncta were increased where they overlapped with actin waves, suggesting a mechanism for the coupling between these two networks. All of these phenotypes could be reversed by inhibiting signal transduction. Strikingly, maintaining cells in this state of constant activation led to a form of cell death by catastrophic fragmentation. These findings provide insight into the feedback loops that control excitability of the signal transduction network, which drives migration.


Asunto(s)
Dictyostelium/fisiología , Proteínas Protozoarias/fisiología , Transducción de Señal/fisiología , Citoesqueleto de Actina/fisiología , Citoesqueleto de Actina/ultraestructura , Adhesión Celular , Movimiento Celular , Forma de la Célula , Quimiotaxis , Dictyostelium/genética , Dictyostelium/ultraestructura , Activación Enzimática , Microscopía Fluorescente , Microscopía de Contraste de Fase , Mutación Missense , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/fisiología , Fenotipo , Proteínas Protozoarias/genética , Proteínas Recombinantes/metabolismo , Proteínas de Unión al GTP rap1/genética , Proteínas de Unión al GTP rap1/fisiología
9.
Proc Natl Acad Sci U S A ; 115(39): E9125-E9134, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30194235

RESUMEN

Signal transduction and cytoskeleton networks in a wide variety of cells display excitability, but the mechanisms are poorly understood. Here, we show that during random migration and in response to chemoattractants, cells maintain complementary spatial and temporal distributions of Ras activity and phosphatidylinositol (3,4)-bisphosphate [PI(3,4)P2]. In addition, depletion of PI(3,4)P2 by disruption of the 5-phosphatase, Dd5P4, or by recruitment of 4-phosphatase INPP4B to the plasma membrane, leads to elevated Ras activity, cell spreading, and altered migratory behavior. Furthermore, RasGAP2 and RapGAP3 bind to PI(3,4)P2, and the phenotypes of cells lacking these genes mimic those with low PI(3,4)P2 levels, providing a molecular mechanism. These findings suggest that Ras activity drives PI(3,4)P2 down, causing the PI(3,4)P2-binding GAPs to dissociate from the membrane, further activating Ras, completing a positive-feedback loop essential for excitability. Consistently, a computational model incorporating such a feedback loop in an excitable network model accurately simulates the dynamic distributions of active Ras and PI(3,4)P2 as well as cell migratory behavior. The mutually inhibitory Ras-PI(3,4)P2 mechanisms we uncovered here provide a framework for Ras regulation that may play a key role in many physiological processes.


Asunto(s)
Membrana Celular/metabolismo , Dictyostelium/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Protozoarias/metabolismo , Transducción de Señal/fisiología , Proteínas ras/metabolismo , Membrana Celular/genética , Dictyostelium/genética , Fosfatos de Fosfatidilinositol/genética , Proteínas Protozoarias/genética , Proteínas ras/genética
10.
Mol Syst Biol ; 15(3): e8585, 2019 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-30858181

RESUMEN

Cellular protrusions are typically considered as distinct structures associated with specific regulators. However, we found that these regulators coordinately localize as propagating cortical waves, suggesting a common underlying mechanism. These molecular events fell into two excitable networks, the signal transduction network STEN and the cytoskeletal network CEN with different wave substructures. Computational studies using a coupled-network model reproduced these features and showed that the morphology and kinetics of the waves depended on strengths of feedback loops. Chemically induced dimerization at multiple nodes produced distinct, coordinated alterations in patterns of other network components. Taken together, these studies indicate: STEN positive feedback is mediated by mutual inhibition between Ras/Rap and PIP2, while negative feedback depends on delayed PKB activation; PKBs link STEN to CEN; CEN includes positive feedback between Rac and F-actin, and exerts fast positive and slow negative feedbacks to STEN The alterations produced protrusions resembling filopodia, ruffles, pseudopodia, or lamellipodia, suggesting that these structures arise from a common regulatory mechanism and that the overall state of the STEN-CEN system determines cellular morphology.


Asunto(s)
Extensiones de la Superficie Celular , Citoesqueleto/metabolismo , Modelos Teóricos , Transducción de Señal , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Simulación por Computador , Microscopía Confocal , Seudópodos , Imagen de Lapso de Tiempo
11.
Proc Natl Acad Sci U S A ; 114(37): E7727-E7736, 2017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28847951

RESUMEN

The model organism Dictyostelium discoideum has greatly facilitated our understanding of the signal transduction and cytoskeletal pathways that govern cell motility. Cell-substrate adhesion is downstream of many migratory and chemotaxis signaling events. Dictyostelium cells lacking the tumor suppressor PTEN show strongly impaired migratory activity and adhere strongly to their substrates. We reasoned that other regulators of migration could be obtained through a screen for overly adhesive mutants. A screen of restriction enzyme-mediated integration mutagenized cells yielded numerous mutants with the desired phenotypes, and the insertion sites in 18 of the strains were mapped. These regulators of adhesion and motility mutants have increased adhesion and decreased motility. Characterization of seven strains demonstrated decreased directed migration, flatness, increased filamentous actin-based protrusions, and increased signal transduction network activity. Many of the genes share homology to human genes and demonstrate the diverse array of cellular networks that function in adhesion and migration.


Asunto(s)
Adhesión Celular/genética , Dictyostelium/genética , Pruebas Genéticas/métodos , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Movimiento Celular/genética , Quimiotaxis/genética , Quimiotaxis/fisiología , AMP Cíclico/metabolismo , Citoesqueleto/metabolismo , Dictyostelium/metabolismo , Proteínas Protozoarias/metabolismo , Resistencia al Corte/fisiología , Transducción de Señal
12.
PLoS Biol ; 14(2): e1002381, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26890004

RESUMEN

For directional movement, eukaryotic cells depend on the proper organization of their actin cytoskeleton. This engine of motility is made up of highly dynamic nonequilibrium actin structures such as flashes, oscillations, and traveling waves. In Dictyostelium, oscillatory actin foci interact with signals such as Ras and phosphatidylinositol 3,4,5-trisphosphate (PIP3) to form protrusions. However, how signaling cues tame actin dynamics to produce a pseudopod and guide cellular motility is a critical open question in eukaryotic chemotaxis. Here, we demonstrate that the strength of coupling between individual actin oscillators controls cell polarization and directional movement. We implement an inducible sequestration system to inactivate the heterotrimeric G protein subunit Gß and find that this acute perturbation triggers persistent, high-amplitude cortical oscillations of F-actin. Actin oscillators that are normally weakly coupled to one another in wild-type cells become strongly synchronized following acute inactivation of Gß. This global coupling impairs sensing of internal cues during spontaneous polarization and sensing of external cues during directional motility. A simple mathematical model of coupled actin oscillators reveals the importance of appropriate coupling strength for chemotaxis: moderate coupling can increase sensitivity to noisy inputs. Taken together, our data suggest that Gß regulates the strength of coupling between actin oscillators for efficient polarity and directional migration. As these observations are only possible following acute inhibition of Gß and are masked by slow compensation in genetic knockouts, our work also shows that acute loss-of-function approaches can complement and extend the reach of classical genetics in Dictyostelium and likely other systems as well.


Asunto(s)
Actinas/metabolismo , Movimiento Celular , Polaridad Celular , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Modelos Biológicos , Relojes Biológicos , Citoesqueleto/metabolismo , Dictyostelium , Transducción de Señal , Sirolimus
13.
Proc Natl Acad Sci U S A ; 113(47): E7500-E7509, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27821730

RESUMEN

Signal transduction pathways activated by chemoattractants have been extensively studied, but little is known about the events mediating responses to mechanical stimuli. We discovered that acute mechanical perturbation of cells triggered transient activation of all tested components of the chemotactic signal transduction network, as well as actin polymerization. Similarly to chemoattractants, the shear flow-induced signal transduction events displayed features of excitability, including the ability to mount a full response irrespective of the length of the stimulation and a refractory period that is shared with that generated by chemoattractants. Loss of G protein subunits, inhibition of multiple signal transduction events, or disruption of calcium signaling attenuated the response to acute mechanical stimulation. Unlike the response to chemoattractants, an intact actin cytoskeleton was essential for reacting to mechanical perturbation. These results taken together suggest that chemotactic and mechanical stimuli trigger activation of a common signal transduction network that integrates external cues to regulate cytoskeletal activity and drive cell migration.


Asunto(s)
Citoesqueleto de Actina/genética , Factores Quimiotácticos/farmacología , Dictyostelium/fisiología , Redes Reguladoras de Genes , Estrés Mecánico , Señalización del Calcio , Movimiento Celular , Citoesqueleto/metabolismo , Dictyostelium/metabolismo , Proteínas de Unión al GTP/metabolismo , Redes Reguladoras de Genes/efectos de los fármacos , Genes Protozoarios , Transducción de Señal
14.
Biochem Biophys Res Commun ; 507(1-4): 304-310, 2018 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-30454895

RESUMEN

The wide range sensing of extracellular signals is a common feature of various sensory cells. Eukaryotic chemotactic cells driven by GPCRs and their cognate G proteins are one example. This system endows the cells directional motility towards their destination over long distances. There are several mechanisms to achieve the long dynamic range, including negative regulation of the receptors upon ligand interaction and spatial regulation of G proteins, as we found recently. However, these mechanisms are insufficient to explain the 105-fold range of chemotaxis seen in Dictyostelium. Here, we reveal that the receptor-mediated activation, recruitment, and capturing of G proteins mediate chemotactic signaling at the lower, middle and higher concentration ranges, respectively. These multiple mechanisms of G protein dynamics can successfully cover distinct ranges of ligand concentrations, resulting in seamless and broad chemotaxis. Furthermore, single-molecule imaging analysis showed that the activated Gα subunit forms an unconventional complex with the agonist-bound receptor. This complex formation of GPCR-Gα increased the membrane-binding time of individual Gα molecules and therefore resulted in the local accumulation of Gα. Our findings provide an additional chemotactic dynamic range mechanism in which multiple G protein dynamics positively contribute to the production of gradient information.


Asunto(s)
Quimiotaxis , Dictyostelium/citología , Dictyostelium/metabolismo , Proteínas de Unión al GTP/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , AMP Cíclico/metabolismo , Espacio Intracelular/metabolismo , Proteínas Protozoarias/metabolismo , Transducción de Señal
15.
Proc Natl Acad Sci U S A ; 112(29): E3845-54, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26130809

RESUMEN

Asymmetric protein localization is essential for cell polarity and migration. We report a novel protein, Callipygian (CynA), which localizes to the lagging edge before other proteins and becomes more tightly restricted as cells polarize; additionally, it accumulates in the cleavage furrow during cytokinesis. CynA protein that is tightly localized, or "clustered," to the cell rear is immobile, but when polarity is disrupted, it disperses throughout the membrane and responds to uniform chemoattractant stimulation by transiently localizing to the cytosol. These behaviors require a pleckstrin homology-domain membrane tether and a WD40 clustering domain, which can also direct other membrane proteins to the back. Fragments of CynA lacking the pleckstrin homology domain, which are normally found in the cytosol, localize to the lagging edge membrane when coexpressed with full-length protein, showing that CynA clustering is mediated by oligomerization. Cells lacking CynA have aberrant lateral protrusions, altered leading-edge morphology, and decreased directional persistence, whereas those overexpressing the protein display exaggerated features of polarity. Consistently, actin polymerization is inhibited at sites of CynA accumulation, thereby restricting protrusions to the opposite edge. We suggest that the mutual antagonism between CynA and regions of responsiveness creates a positive feedback loop that restricts CynA to the rear and contributes to the establishment of the cell axis.


Asunto(s)
Movimiento Celular , Polaridad Celular , Dictyostelium/citología , Proteínas Protozoarias/metabolismo , Actinas/metabolismo , Agregación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Extensiones de la Superficie Celular/efectos de los fármacos , Extensiones de la Superficie Celular/metabolismo , Factores Quimiotácticos/farmacología , Dictyostelium/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Fosfatidilinositoles/farmacología , Polimerizacion/efectos de los fármacos , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Proteínas Protozoarias/química , Transducción de Señal/efectos de los fármacos
16.
Proc Natl Acad Sci U S A ; 111(26): E2684-93, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24979808

RESUMEN

The signaling lipid phosphatidylinositol (3,4,5)-trisphosphate (PIP3) is a key regulator of cell proliferation, survival, and migration and the enzyme that dephosphorylates it, phosphatase and tensin homolog (PTEN), is an important tumor suppressor. As excess PIP3 signaling is a hallmark of many cancers, its suppression through activation of PTEN is a potential cancer intervention. Using a heterologous expression system in which human PTEN-GFP is expressed in Dictyostelium cells, we identified mutations in the membrane-binding regulatory interface that increase the recruitment of PTEN to the plasma membrane due to enhanced association with PI(4,5)P2. We engineered these into an enhanced PTEN (ePTEN) with approximately eightfold increased ability to suppress PIP3 signaling. Upon expression in human cells, ePTEN decreases PIP3 levels in the plasma membrane; phosphorylation of AKT, a major downstream event in PIP3 signaling; and cell proliferation and migration. Thus, the activation of PTEN can readjust PIP3 signaling and may serve as a feasible target for anticancer therapies.


Asunto(s)
Fosfohidrolasa PTEN/genética , Fosfatos de Fosfatidilinositol/antagonistas & inhibidores , Ingeniería de Proteínas/métodos , Proteínas Recombinantes/genética , Transducción de Señal/efectos de los fármacos , Proteínas Supresoras de Tumor/genética , Células Cultivadas , Clonación Molecular , Dictyostelium , Biblioteca de Genes , Proteínas Fluorescentes Verdes , Células HEK293 , Humanos , Immunoblotting , Mutagénesis , Proteínas Recombinantes/farmacología
17.
Methods ; 77-78: 119-24, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25448479

RESUMEN

Many tumors are associated with deficiency of the tumor suppressor, PTEN, a PIP3 phosphatase that turns off PIP3 signaling. The major site of PTEN action is the plasma membrane, where PIP3 is produced by PI3 kinases. However, the mechanism and functional importance of PTEN membrane recruitment are poorly defined. Using the heterologous expression system in which human PTEN is expressed in Dictyostelium discoideum, we defined the molecular mechanisms that regulate the membrane-binding site through inhibitory interactions with the phosphorylated C-terminal tail. In addition, we potentiated mechanisms that mediate PTEN membrane association and engineered an enhanced PTEN with increased tumor suppressor functions. Moreover, we identified a new class of cancer-associated PTEN mutations that are specifically defective in membrane association. In this review, we summarize recent advances in PTEN-membrane interactions and methods useful in addressing PTEN function.


Asunto(s)
Membrana Celular/metabolismo , Dictyostelium/metabolismo , Ingeniería Genética/métodos , Fosfohidrolasa PTEN/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Membrana Celular/genética , Dictyostelium/genética , Células HEK293 , Humanos , Fosfohidrolasa PTEN/química , Fosfohidrolasa PTEN/genética , Estructura Secundaria de Proteína , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética
18.
Curr Opin Cell Biol ; 20(1): 35-40, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18207721

RESUMEN

Chemotaxis in eukaryotic cells involves the coordination of several related but separable processes: motility, polarization, and gradient sensing. Mathematical models that have been proposed to explain chemotaxis typically focus on only one of these processes. We summarize the strengths and weaknesses of the models and point out the need for an integrated model.


Asunto(s)
Quimiotaxis , Modelos Biológicos , Animales , Polaridad Celular
19.
Cell Mol Life Sci ; 71(19): 3711-47, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24846395

RESUMEN

Chemotaxis, or directed migration of cells along a chemical gradient, is a highly coordinated process that involves gradient sensing, motility, and polarity. Most of our understanding of chemotaxis comes from studies of cells undergoing amoeboid-type migration, in particular the social amoeba Dictyostelium discoideum and leukocytes. In these amoeboid cells the molecular events leading to directed migration can be conceptually divided into four interacting networks: receptor/G protein, signal transduction, cytoskeleton, and polarity. The signal transduction network occupies a central position in this scheme as it receives direct input from the receptor/G protein network, as well as feedback from the cytoskeletal and polarity networks. Multiple overlapping modules within the signal transduction network transmit the signals to the actin cytoskeleton network leading to biased pseudopod protrusion in the direction of the gradient. The overall architecture of the networks, as well as the individual signaling modules, is remarkably conserved between Dictyostelium and mammalian leukocytes, and the similarities and differences between the two systems are the subject of this review.


Asunto(s)
Dictyostelium/metabolismo , Leucocitos/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Adhesión Celular/fisiología , Proteínas de Ciclo Celular/metabolismo , Quimiotaxis de Leucocito/fisiología , Leucocitos/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
20.
Proc Natl Acad Sci U S A ; 109(34): 13632-7, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22847424

RESUMEN

Chemotaxis depends on a network of parallel pathways that coordinate cytoskeletal events to bias cell movement along a chemoattractant gradient. Using a forward genetic screen in Dictyostelium discoideum, we identified the Ste20 kinase KrsB, a homolog of tumor suppressors Hippo and MST1/2, as a negative regulator of cell spreading and substrate attachment. The excessive adhesion of krsB(-) cells reduced directional movement and prolonged the streaming phase of multicellular aggregation. These phenotypes depended on an intact kinase domain and phosphorylation of a conserved threonine (T176) within the activation loop. Chemoattractants triggered a rapid, transient autophosphorylation of T176 in a heterotrimeric G protein-dependent and PI3K- and TorC2-independent manner. The active phosphorylated form of KrsB acts to decrease adhesion to the substrate. Taken together these studies suggest that cycling between active and inactive forms of KrsB may provide the dynamic regulation of cell adhesion needed for proper cell migration and chemotaxis. KrsB interacts genetically with another D. discoideum Hippo/MST homolog, KrsA, but the two genes are not functionally redundant. These studies show that Hippo/MST proteins, like the tumor suppressor PTEN and oncogenes Ras and PI3K, play a key role in cell morphological events in addition to their role in regulating cell growth.


Asunto(s)
Quimiotaxis , Regulación Neoplásica de la Expresión Génica , Factor de Crecimiento de Hepatocito/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Protozoarias/genética , Animales , Adhesión Celular , Movimiento Celular , Dictyostelium , Dimerización , Genes Supresores de Tumor , Proteínas Fluorescentes Verdes/química , Factor de Crecimiento de Hepatocito/química , Humanos , Proteínas del Tejido Nervioso/química , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/química , Proteínas Proto-Oncogénicas/química , Treonina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA