Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Cell Biol ; 26(7): 1062-1076, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38951708

RESUMO

Ras has been extensively studied as a promoter of cell proliferation, whereas few studies have explored its role in migration. To investigate the direct and immediate effects of Ras activity on cell motility or polarity, we focused on RasGAPs, C2GAPB in Dictyostelium amoebae and RASAL3 in HL-60 neutrophils and macrophages. In both cellular systems, optically recruiting the respective RasGAP to the cell front extinguished pre-existing protrusions and changed migration direction. However, when these respective RasGAPs were recruited uniformly to the membrane, cells polarized and moved more rapidly, whereas targeting to the back exaggerated these effects. These unexpected outcomes of attenuating Ras activity naturally had strong, context-dependent consequences for chemotaxis. The RasGAP-mediated polarization depended critically on myosin II activity and commenced with contraction at the cell rear, followed by sustained mTORC2-dependent actin polymerization at the front. These experimental results were captured by computational simulations in which Ras levels control front- and back-promoting feedback loops. The discovery that inhibiting Ras activity can produce counterintuitive effects on cell migration has important implications for future drug-design strategies targeting oncogenic Ras.


Assuntos
Actomiosina , Movimento Celular , Polaridade Celular , Dictyostelium , Proteínas ras , Dictyostelium/metabolismo , Dictyostelium/genética , Células HL-60 , Actomiosina/metabolismo , Humanos , Proteínas ras/metabolismo , Proteínas ras/genética , Macrófagos/metabolismo , Miosina Tipo II/metabolismo , Miosina Tipo II/genética , Neutrófilos/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo , Proteínas Ativadoras de ras GTPase/genética , Animais , Quimiotaxia , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/genética , Actinas/metabolismo , Simulação por Computador , Camundongos , Transdução de Sinais
2.
bioRxiv ; 2023 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-37693515

RESUMO

Studies in the model systems, Dictyostelium amoebae and HL-60 neutrophils, have shown that local Ras activity directly regulates cell motility or polarity. Localized Ras activation on the membrane is spatiotemporally regulated by its activators, RasGEFs, and inhibitors, RasGAPs, which might be expected to create a stable 'front' and 'back', respectively, in migrating cells. Focusing on C2GAPB in amoebae and RASAL3 in neutrophils, we investigated how Ras activity along the cortex controls polarity. Since existing gene knockout and overexpression studies can be circumvented, we chose optogenetic approaches to assess the immediate, local effects of these Ras regulators on the cell cortex. In both cellular systems, optically targeting the respective RasGAPs to the cell front extinguished existing protrusions and changed the direction of migration, as might be expected. However, when the expression of C2GAPB was induced globally, amoebae polarized within hours. Furthermore, within minutes of globally recruiting either C2GAPB in amoebae or RASAL3 in neutrophils, each cell type polarized and moved more rapidly. Targeting the RasGAPs to the cell backs exaggerated these effects on migration and polarity. Overall, in both cell types, RasGAP-mediated polarization was brought about by increased actomyosin contractility at the back and sustained, localized F-actin polymerization at the front. These experimental results were accurately captured by computational simulations in which Ras levels control front and back feedback loops. The discovery that context-dependent Ras activity on the cell cortex has counterintuitive, unanticipated effects on cell polarity can have important implications for future drug-design strategies targeting oncogenic Ras.

3.
Dev Cell ; 58(13): 1170-1188.e7, 2023 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-37220748

RESUMO

Ras signaling is typically associated with cell growth, but not direct regulation of motility or polarity. By optogenetically targeting different nodes in the Ras/PI3K/Akt network in differentiated human HL-60 neutrophils, we abruptly altered protrusive activity, bypassing the chemoattractant receptor/G-protein network. First, global recruitment of active KRas4B/HRas isoforms or a RasGEF, RasGRP4, immediately increased spreading and random motility. Second, activating Ras at the cell rear generated new protrusions, reversed pre-existing polarity, and steered sustained migration in neutrophils or murine RAW 264.7 macrophages. Third, recruiting a RasGAP, RASAL3, to cell fronts extinguished protrusions and changed migration direction. Remarkably, persistent RASAL3 recruitment at stable fronts abrogated directed migration in three different chemoattractant gradients. Fourth, local recruitment of the Ras-mTORC2 effector, Akt, in neutrophils or Dictyostelium amoebae generated new protrusions and rearranged pre-existing polarity. Overall, these optogenetic effects were mTORC2-dependent but relatively independent of PI3K. Thus, receptor-independent, local activations of classical growth-control pathways directly control actin assembly, cell shape, and migration modes.


Assuntos
Dictyostelium , Proteínas Proto-Oncogênicas c-akt , Animais , Camundongos , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Dictyostelium/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Movimento Celular/fisiologia , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular
4.
Nat Cell Biol ; 24(10): 1499-1515, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36202973

RESUMO

During cell migration and polarization, numerous signal transduction and cytoskeletal components self-organize to generate localized protrusions. Although biochemical and genetic analyses have delineated many specific interactions, how the activation and localization of so many different molecules are spatiotemporally orchestrated at the subcellular level has remained unclear. Here we show that the regulation of negative surface charge on the inner leaflet of the plasma membrane plays an integrative role in the molecular interactions. Surface charge, or zeta potential, is transiently lowered at new protrusions and within cortical waves of Ras/PI3K/TORC2/F-actin network activation. Rapid alterations of inner leaflet anionic phospholipids-such as PI(4,5)P2, PI(3,4)P2, phosphatidylserine and phosphatidic acid-collectively contribute to the surface charge changes. Abruptly reducing the surface charge by recruiting positively charged optogenetic actuators was sufficient to trigger the entire biochemical network, initiate de novo protrusions and abrogate pre-existing polarity. These effects were blocked by genetic or pharmacological inhibition of key signalling components such as AKT and PI3K/TORC2. Conversely, increasing the negative surface charge deactivated the network and locally suppressed chemoattractant-induced protrusions or subverted EGF-induced ERK activation. Computational simulations involving excitable biochemical networks demonstrated that slight changes in feedback loops, induced by recruitment of the charged actuators, could lead to outsized effects on system activation. We propose that key signalling network components act on, and are in turn acted upon, by surface charge, closing feedback loops, which bring about the global-scale molecular self-organization required for spontaneous protrusion formation, cell migration and polarity establishment.


Assuntos
Actinas , Polaridade Celular , Polaridade Celular/fisiologia , Actinas/metabolismo , Fosfatidilserinas/metabolismo , Fator de Crescimento Epidérmico , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Movimento Celular , Membrana Celular/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Fatores Quimiotáticos/metabolismo
5.
Biophys J ; 121(23): 4600-4614, 2022 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-36273263

RESUMO

Cell shape change processes, such as proliferation, polarization, migration, and cancer metastasis, rely on a dynamic network of macromolecules. The proper function of this network enables mechanosensation, the ability of cells to sense and respond to mechanical cues. Myosin II and cortexillin I, critical elements of the cellular mechanosensory machinery, preassemble in the cytoplasm of Dictyostelium cells into complexes that we have termed contractility kits (CKs). Two IQGAP proteins then differentially regulate the mechanoresponsiveness of the cortexillin I-myosin II elements within CKs. To investigate the mechanism of CK self-assembly and gain insight into possible molecular means for IQGAP regulation, we developed a coarse-grained excluded volume molecular model in which all protein polymers are represented by nm-sized spheres connected by spring-like links. The model is parameterized using experimentally measured parameters acquired through fluorescence cross-correlation spectroscopy and fluorescence correlation spectroscopy, which describe the interaction affinities and diffusion coefficients for individual molecular components, and which have also been validated via several orthogonal methods. Simulations of wild-type and null-mutant conditions implied that the temporal order of assembly of these kits is dominated by myosin II dimer formation and that IQGAP proteins mediate cluster growth. In addition, our simulations predicted the existence of "ambiguous" CKs that incorporate both classes of IQGAPs, and we confirmed this experimentally using fluorescence cross-correlation spectroscopy. The model serves to describe the formation of the CKs and how their assembly enables and regulates mechanosensation at the molecular level.


Assuntos
Dictyostelium
6.
Cell ; 184(25): 6193-6206.e14, 2021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34838160

RESUMO

Genetically encoded fluorescent biosensors are powerful tools for monitoring biochemical activities in live cells, but their multiplexing capacity is limited by the available spectral space. We overcome this problem by developing a set of barcoding proteins that can generate over 100 barcodes and are spectrally separable from commonly used biosensors. Mixtures of barcoded cells expressing different biosensors are simultaneously imaged and analyzed by deep learning models to achieve massively multiplexed tracking of signaling events. Importantly, different biosensors in cell mixtures show highly coordinated activities, thus facilitating the delineation of their temporal relationship. Simultaneous tracking of multiple biosensors in the receptor tyrosine kinase signaling network reveals distinct mechanisms of effector adaptation, cell autonomous and non-autonomous effects of KRAS mutations, as well as complex interactions in the network. Biosensor barcoding presents a scalable method to expand multiplexing capabilities for deciphering the complexity of signaling networks and their interactions between cells.


Assuntos
Técnicas Biossensoriais/métodos , Células/ultraestrutura , Microscopia de Fluorescência/métodos , Análise de Célula Única/métodos , Linhagem Celular Tumoral , Humanos
7.
Cell Rep ; 34(12): 108892, 2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33761356

RESUMO

Most solid tumors are aneuploid, and p53 has been implicated as the guardian of the euploid genome. Previous experiments using human cell lines showed that aneuploidy induction leads to p53 accumulation and p21-mediated G1 cell cycle arrest. We find that adherent 2-dimensional (2D) cultures of human immortalized or cancer cell lines activate p53 upon aneuploidy induction, whereas suspension cultures of a human lymphoid cell line undergo a p53-independent cell cycle arrest. Surprisingly, 3D human and mouse organotypic cultures from neural, intestinal, or mammary epithelial tissues do not activate p53 or arrest in G1 following aneuploidy induction. p53-deficient colon organoids have increased aneuploidy and frequent lagging chromosomes and multipolar spindles during mitosis. These data suggest that p53 may not act as a universal surveillance factor restricting the proliferation of aneuploid cells but instead helps directly or indirectly ensure faithful chromosome transmission likely by preventing polyploidization and influencing spindle mechanics.


Assuntos
Aneuploidia , Proteína Supressora de Tumor p53/metabolismo , Animais , Adesão Celular , Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Colo/metabolismo , Humanos , Mamíferos , Camundongos Endogâmicos C57BL , Mitose , Organoides/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo
8.
EMBO J ; 40(4): e105094, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33586225

RESUMO

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.


Assuntos
Membrana Celular/metabolismo , Movimento Celular , Dictyostelium/fisiologia , Microtúbulos/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Dictyostelium/citologia , Células HL-60 , Humanos
9.
J Cell Sci ; 134(4)2021 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-33526710

RESUMO

Airway hydration and ciliary function are critical to airway homeostasis and dysregulated in chronic obstructive pulmonary disease (COPD), which is impacted by cigarette smoking and has no therapeutic options. We utilized a high-copy cDNA library genetic selection approach in the amoeba Dictyostelium discoideum to identify genetic protectors to cigarette smoke. Members of the mitochondrial ADP/ATP transporter family adenine nucleotide translocase (ANT) are protective against cigarette smoke in Dictyostelium and human bronchial epithelial cells. Gene expression of ANT2 is reduced in lung tissue from COPD patients and in a mouse smoking model, and overexpression of ANT1 and ANT2 resulted in enhanced oxidative respiration and ATP flux. In addition to the presence of ANT proteins in the mitochondria, they reside at the plasma membrane in airway epithelial cells and regulate airway homeostasis. ANT2 overexpression stimulates airway surface hydration by ATP and maintains ciliary beating after exposure to cigarette smoke, both of which are key functions of the airway. Our study highlights a potential for upregulation of ANT proteins and/or of their agonists in the protection from dysfunctional mitochondrial metabolism, airway hydration and ciliary motility in COPD.This article has an associated First Person interview with the first author of the paper.


Assuntos
Dictyostelium , Doença Pulmonar Obstrutiva Crônica , Dictyostelium/genética , Células Epiteliais/metabolismo , Humanos , Pulmão , Mitocôndrias , Translocases Mitocondriais de ADP e ATP/metabolismo , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/metabolismo
10.
Artigo em Inglês | MEDLINE | ID: mdl-33305503

RESUMO

A cell's ability to change shape is one of the most fundamental biological processes and is essential for maintaining healthy organisms. When the ability to control shape goes awry, it often results in a diseased system. As such, it is important to understand the mechanisms that allow a cell to sense and respond to its environment so as to maintain cellular shape homeostasis. Because of the inherent complexity of the system, computational models that are based on sound theoretical understanding of the biochemistry and biomechanics and that use experimentally measured parameters are an essential tool. These models involve an inherent feedback, whereby shape is determined by the action of regulatory signals whose spatial distribution depends on the shape. To carry out computational simulations of these moving boundary problems requires special computational techniques. A variety of alternative approaches, depending on the type and scale of question being asked, have been used to simulate various biological processes, including cell motility, division, mechanosensation, and cell engulfment. In general, these models consider the forces that act on the system (both internally generated, or externally imposed) and the mechanical properties of the cell that resist these forces. Moving forward, making these techniques more accessible to the non-expert will help improve interdisciplinary research thereby providing new insight into important biological processes that affect human health. This article is categorized under: Cancer > Cancer>Computational Models Cancer > Cancer>Molecular and Cellular Physiology.

11.
Dev Cell ; 54(5): 608-623.e5, 2020 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-32877650

RESUMO

The Ras/PI3K/extracellular signal-regulated kinases (ERK) signaling network plays fundamental roles in cell growth, survival, and migration and is frequently activated in cancer. Here, we show that the activities of the signaling network propagate as coordinated waves, biased by growth factor, which drive actin-based protrusions in human epithelial cells. The network exhibits hallmarks of biochemical excitability: the annihilation of oppositely directed waves, all-or-none responsiveness, and refractoriness. Abrupt perturbations to Ras, PI(4,5)P2, PI(3,4)P2, ERK, and TORC2 alter the threshold, observations that define positive and negative feedback loops within the network. Oncogenic transformation dramatically increases the wave activity, the frequency of ERK pulses, and the sensitivity to EGF stimuli. Wave activity was progressively enhanced across a series of increasingly metastatic breast cancer cell lines. The view that oncogenic transformation is a shift to a lower threshold of excitable Ras/PI3K/ERK network, caused by various combinations of genetic insults, can facilitate the assessment of cancer severity and effectiveness of interventions.


Assuntos
Carcinogênese/patologia , Transformação Celular Neoplásica/genética , Células Epiteliais/metabolismo , Transdução de Sinais/fisiologia , Actinas/metabolismo , Carcinogênese/genética , Linhagem Celular Tumoral , Transformação Celular Neoplásica/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética , Proteínas ras/metabolismo
12.
Cancer Res ; 79(18): 4665-4678, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31358530

RESUMO

Metastasis is complex, involving multiple genetic, epigenetic, biochemical, and physical changes in the cancer cell and its microenvironment. Cells with metastatic potential are often characterized by altered cellular contractility and deformability, lending them the flexibility to disseminate and navigate through different microenvironments. We demonstrate that mechanoresponsiveness is a hallmark of pancreatic cancer cells. Key mechanoresponsive proteins, those that accumulate in response to mechanical stress, specifically nonmuscle myosin IIA (MYH9) and IIC (MYH14), α-actinin 4, and filamin B, were highly expressed in pancreatic cancer as compared with healthy ductal epithelia. Their less responsive sister paralogs-myosin IIB (MYH10), α-actinin 1, and filamin A-had lower expression differential or disappeared with cancer progression. We demonstrate that proteins whose cellular contributions are often overlooked because of their low abundance can have profound impact on cell architecture, behavior, and mechanics. Here, the low abundant protein MYH14 promoted metastatic behavior and could be exploited with 4-hydroxyacetophenone (4-HAP), which increased MYH14 assembly, stiffening cells. As a result, 4-HAP decreased dissemination, induced cortical actin belts in spheroids, and slowed retrograde actin flow. 4-HAP also reduced liver metastases in human pancreatic cancer-bearing nude mice. Thus, increasing MYH14 assembly overwhelms the ability of cells to polarize and invade, suggesting targeting the mechanoresponsive proteins of the actin cytoskeleton as a new strategy to improve the survival of patients with pancreatic cancer. SIGNIFICANCE: This study demonstrates that mechanoresponsive proteins become upregulated with pancreatic cancer progression and that this system of proteins can be pharmacologically targeted to inhibit the metastatic potential of pancreatic cancer cells.


Assuntos
Acetofenonas/farmacologia , Actinina/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo II/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Citoesqueleto de Actina , Actinina/genética , Animais , Apoptose , Proliferação de Células , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundário , Camundongos , Camundongos Nus , Cadeias Pesadas de Miosina/genética , Miosina Tipo II/genética , Invasividade Neoplásica , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Prognóstico , Células Tumorais Cultivadas , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Nat Commun ; 9(1): 4673, 2018 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-30405112

RESUMO

The Ras-ERK signaling pathway regulates diverse cellular processes in response to environmental stimuli and contains important therapeutic targets for cancer. Recent single cell studies revealed stochastic pulses of ERK activation, the frequency of which determines functional outcomes such as cell proliferation. Here we show that ERK pulses are initiated by localized protrusive activities. Chemically and optogenetically induced protrusions trigger ERK activation through various entry points into the feedback loop involving Ras, PI3K, the cytoskeleton, and cellular adhesion. The excitability of the protrusive signaling network drives stochastic ERK activation in unstimulated cells and oscillations upon growth factor stimulation. Importantly, protrusions allow cells to sense combined signals from substrate stiffness and the growth factor. Thus, by uncovering the basis of ERK pulse generation we demonstrate how signals involved in cell growth and differentiation are regulated by dynamic protrusions that integrate chemical and mechanical inputs from the environment.


Assuntos
Extensões da Superfície Celular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Mecanotransdução Celular , Linhagem Celular , Linhagem Celular Tumoral , Simulação por Computador , Ativação Enzimática , Humanos , Modelos Biológicos
14.
Proc Natl Acad Sci U S A ; 115(39): E9125-E9134, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-30194235

RESUMO

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.


Assuntos
Membrana Celular/metabolismo , Dictyostelium/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas de Protozoários/metabolismo , Transdução de Sinais/fisiologia , Proteínas ras/metabolismo , Membrana Celular/genética , Dictyostelium/genética , Fosfatos de Fosfatidilinositol/genética , Proteínas de Protozoários/genética , Proteínas ras/genética
15.
Annu Rev Cell Dev Biol ; 33: 103-125, 2017 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-28793794

RESUMO

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.


Assuntos
Movimento Celular , Transdução de Sinais , Animais , Humanos , Modelos Biológicos
16.
Nat Cell Biol ; 19(4): 329-340, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28346441

RESUMO

The diverse migratory modes displayed by different cell types are generally believed to be idiosyncratic. Here we show that the migratory behaviour of Dictyostelium was switched from amoeboid to keratocyte-like and oscillatory modes by synthetically decreasing phosphatidylinositol-4,5-bisphosphate levels or increasing Ras/Rap-related activities. The perturbations at these key nodes of an excitable signal transduction network initiated a causal chain of events: the threshold for network activation was lowered, the speed and range of propagating waves of signal transduction activity increased, actin-driven cellular protrusions expanded and, consequently, the cell migratory mode transitions ensued. Conversely, innately keratocyte-like and oscillatory cells were promptly converted to amoeboid by inhibition of Ras effectors with restoration of directed migration. We use computational analysis to explain how thresholds control cell migration and discuss the architecture of the signal transduction network that gives rise to excitability.


Assuntos
Movimento Celular , Dictyostelium/citologia , Dictyostelium/metabolismo , Transdução de Sinais , Actinas/metabolismo , Técnicas Biossensoriais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Simulação por Computador , AMP Cíclico/farmacologia , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Dictyostelium/efeitos dos fármacos , Proteínas de Fluorescência Verde/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Imagem com Lapso de Tempo
17.
Cell Rep ; 9(3): 1110-21, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25437564

RESUMO

Polarized migrating cells display signal transduction events, such as activation of phosphatidylinositol 3-kinase (PI3K) and Scar/Wave, and respond more readily to chemotactic stimuli at the leading edge. We sought to determine the basis of this polarized sensitivity. Inhibiting actin polymerization leads to uniform sensitivity. However, when human neutrophils were "stalled" by simultaneously blocking actin and myosin dynamics, they maintained the gradient of responsiveness to chemoattractant and also displayed noise-driven PIP3 flashes on the basal membrane, localized toward the front. Thus, polarized sensitivity does not require migration or cytoskeletal dynamics. The threshold for response is correlated with the static F-actin distribution, but not cell shape or volume changes, membrane fluidity, or the preexisting distribution of PI3K. The kinetics of responses to temporal and spatial stimuli were consistent with the local excitation global inhibition model, but the overall direction of the response was biased by the internal axis of polarity.


Assuntos
Polaridade Celular/efeitos dos fármacos , Fatores Quimiotáticos/farmacologia , Quimiotaxia/efeitos dos fármacos , Citoesqueleto/metabolismo , Neutrófilos/citologia , Actinas/metabolismo , Amidas/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Forma Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Células Imobilizadas/citologia , Células Imobilizadas/efeitos dos fármacos , Células Imobilizadas/metabolismo , Citoesqueleto/efeitos dos fármacos , Depsipeptídeos/farmacologia , Células HL-60 , Humanos , Cinética , Fluidez de Membrana/efeitos dos fármacos , Microscopia de Fluorescência , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-akt/química , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridinas/farmacologia , Tiazolidinas/farmacologia
18.
Science ; 343(6177): 1249531, 2014 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24653039

RESUMO

Biological oscillations are observed at many levels of cellular organization. In the social amoeba Dictyostelium discoideum, starvation-triggered multicellular development is organized by periodic cyclic adenosine 3',5'-monophosphate (cAMP) waves, which provide both chemoattractant gradients and developmental signals. We report that GtaC, a GATA transcription factor, exhibits rapid nucleocytoplasmic shuttling in response to cAMP waves. This behavior requires coordinated action of a nuclear localization signal and reversible G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptor-mediated phosphorylation. Although both are required for developmental gene expression, receptor occupancy promotes nuclear exit of GtaC, which leads to a transient burst of transcription at each cAMP cycle. We demonstrate that this biological circuit filters out high-frequency signals and counts those admitted, thereby enabling cells to modulate gene expression according to the dynamic pattern of the external stimuli.


Assuntos
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Dictyostelium/metabolismo , Fatores de Transcrição GATA/metabolismo , Proteínas de Protozoários/metabolismo , Transporte Ativo do Núcleo Celular , AMP Cíclico/metabolismo , AMP Cíclico/farmacologia , Dictyostelium/crescimento & desenvolvimento , Fatores de Transcrição GATA/química , Fatores de Transcrição GATA/genética , Regulação da Expressão Gênica , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Sinais de Localização Nuclear , Fosforilação , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Receptores Acoplados a Proteínas G/metabolismo
19.
Artigo em Inglês | MEDLINE | ID: mdl-20835994

RESUMO

Phosphoinositide 3-kinases (PI3Ks) are a family of lipid kinases that phosphorylate the 3' OH position of the inositol ring of phosphoinositides on the inner leaf of the plasma membrane. Receptor-mediated activation of the PI3K pathway plays a crucial role in numerous signaling pathways and regulates a number of critical cellular processes, including growth, differentiation, survival and directed migration. In this focus article, we review the temporal and spatial regulation of PI3K in chemotaxing cells with particular emphasis on the amoeba Dictyostelium as well as neutrophils. We also briefly discuss one model used to elucidate the PI3K pathway.


Assuntos
Quimiotaxia/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Animais , Fatores Quimiotáticos/fisiologia , Dictyostelium/fisiologia , Humanos , Modelos Biológicos , Neutrófilos/fisiologia , PTEN Fosfo-Hidrolase/fisiologia , Fosfatidilinositóis/fisiologia , Transdução de Sinais/fisiologia , Biologia de Sistemas
20.
Genes Dev ; 22(16): 2278-90, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18708585

RESUMO

In a forward genetic screen for chemotaxis mutants in Dictyostelium discoideum, we identified a loss-of-function mutation, designated tsunami, encoding a homolog of the Fused kinase. Cells lacking tsuA function could not effectively perform chemotaxis and were unable to become polarized or correctly orient pseudopods in chemotactic gradients. While tsuA(-) cells were able to couple receptor occupancy to phosphatidylinositol (3,4,5) trisphosphate (PIP3) production and actin polymerization, the PIP3 response was prolonged and basal F-actin levels were increased. Interestingly, TsuA localizes to the microtubule network and puncta mainly found at the cell periphery. Analysis of the gene uncovered a novel C-terminal domain that we designated the Tsunami Homology (TH) domain. Both the kinase domain and the TH domain are required to rescue the phenotypic defects of tsuA(-) cells. While kinase activity is not required for localization to microtubules, the TH domain is essential. Thus, localization of kinase activity to microtubules is critical for TsuA function. We propose that functions in association with the microtubule network may underlie the divergent roles of Fused kinase proteins in different organisms.


Assuntos
Movimento Celular , Polaridade Celular , Quimiotaxia/fisiologia , Dictyostelium/fisiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Actinas/metabolismo , Sequência de Aminoácidos , Animais , Diferenciação Celular , AMP Cíclico/metabolismo , Perfilação da Expressão Gênica , Microtúbulos/metabolismo , Dados de Sequência Molecular , Mutação/genética , Fenótipo , Fosfatidilinositóis/metabolismo , Transporte Proteico , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Frações Subcelulares
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA