Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Proc Natl Acad Sci U S A ; 114(47): E10092-E10101, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29109256

RESUMEN

Eukaryotic cells chemotax in a wide range of chemoattractant concentration gradients, and thus need inhibitory processes that terminate cell responses to reach adaptation while maintaining sensitivity to higher-concentration stimuli. However, the molecular mechanisms underlying inhibitory processes are still poorly understood. Here, we reveal a locally controlled inhibitory process in a GPCR-mediated signaling network for chemotaxis in Dictyostelium discoideum We identified a negative regulator of Ras signaling, C2GAP1, which localizes at the leading edge of chemotaxing cells and is activated by and essential for GPCR-mediated Ras signaling. We show that both C2 and GAP domains are required for the membrane targeting of C2GAP1, and that GPCR-triggered Ras activation is necessary to recruit C2GAP1 from the cytosol and retains it on the membrane to locally inhibit Ras signaling. C2GAP1-deficient c2gapA- cells have altered Ras activation that results in impaired gradient sensing, excessive polymerization of F actin, and subsequent defective chemotaxis. Remarkably, these cellular defects of c2gapA- cells are chemoattractant concentration dependent. Thus, we have uncovered an inhibitory mechanism required for adaptation and long-range chemotaxis.


Asunto(s)
Quimiotaxis/genética , Dictyostelium/metabolismo , Proteínas Activadoras de GTPasa/genética , Proteínas Protozoarias/genética , Proteínas ras/genética , Actinas/genética , Actinas/metabolismo , Adaptación Fisiológica , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Quimiotaxis/efectos de los fármacos , AMP Cíclico/metabolismo , AMP Cíclico/farmacología , Citosol/efectos de los fármacos , Citosol/metabolismo , Dictyostelium/efectos de los fármacos , Dictyostelium/genética , Dictyostelium/ultraestructura , Proteínas Activadoras de GTPasa/deficiencia , Regulación de la Expresión Génica , Transporte de Proteínas , Proteínas Protozoarias/metabolismo , Transducción de Señal , Proteínas ras/metabolismo
2.
PLoS Biol ; 12(10): e1001966, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25313567

RESUMEN

The high mortality of melanoma is caused by rapid spread of cancer cells, which occurs unusually early in tumour evolution. Unlike most solid tumours, thickness rather than cytological markers or differentiation is the best guide to metastatic potential. Multiple stimuli that drive melanoma cell migration have been described, but it is not clear which are responsible for invasion, nor if chemotactic gradients exist in real tumours. In a chamber-based assay for melanoma dispersal, we find that cells migrate efficiently away from one another, even in initially homogeneous medium. This dispersal is driven by positive chemotaxis rather than chemorepulsion or contact inhibition. The principal chemoattractant, unexpectedly active across all tumour stages, is the lipid agonist lysophosphatidic acid (LPA) acting through the LPA receptor LPAR1. LPA induces chemotaxis of remarkable accuracy, and is both necessary and sufficient for chemotaxis and invasion in 2-D and 3-D assays. Growth factors, often described as tumour attractants, cause negligible chemotaxis themselves, but potentiate chemotaxis to LPA. Cells rapidly break down LPA present at substantial levels in culture medium and normal skin to generate outward-facing gradients. We measure LPA gradients across the margins of melanomas in vivo, confirming the physiological importance of our results. We conclude that LPA chemotaxis provides a strong drive for melanoma cells to invade outwards. Cells create their own gradients by acting as a sink, breaking down locally present LPA, and thus forming a gradient that is low in the tumour and high in the surrounding areas. The key step is not acquisition of sensitivity to the chemoattractant, but rather the tumour growing to break down enough LPA to form a gradient. Thus the stimulus that drives cell dispersal is not the presence of LPA itself, but the self-generated, outward-directed gradient.


Asunto(s)
Movimiento Celular , Quimiotaxis , Lisofosfolípidos/metabolismo , Melanoma/metabolismo , Metástasis de la Neoplasia , Animales , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones
3.
Biochem Soc Trans ; 43(1): 129-32, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25619258

RESUMEN

The cytoskeleton is utilized for a variety of cellular processes, including migration, endocytosis and adhesion. The required molecular components are often shared between different processes, but it is not well understood how the cells balance their use. We find that macropinocytosis and cell migration are negatively correlated. Heavy drinkers move only slowly and vice versa, fast cells do not take big gulps. Both processes are balanced by the lipid phosphatidylinositol 3,4,5-trisphosphate (PIP3). Elevated PIP3 signalling causes a shift towards macropinocytosis and inhibits motility by redirecting the SCAR/WAVE complex, a major nucleator of actin filaments. High resolution microscopy shows that patches with high levels of PIP3 recruit SCAR/WAVE on their periphery, resulting in circular ruffle formation and engulfment. Results shed new light on the role of PIP3, which is commonly thought to promote cell motility.


Asunto(s)
Movimiento Celular , Pinocitosis , Animales , Dictyostelium/citología , Dictyostelium/fisiología , Humanos
4.
J Cell Sci ; 125(Pt 10): 2457-65, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22366457

RESUMEN

Dock (dedicator of cytokinesis) proteins represent a family of guanine nucleotide exchange factors (GEFs) that include the well-studied Dock180 family and the poorly characterised zizimin family. Our current understanding of Dock180 function is that it regulates Rho small GTPases and thus has a role in a number of cell processes, including cell migration, development and division. Here, we use a tractable model for cell motility research, Dictyostelium discoideum, to help elucidate the role of the related zizimin proteins. We show that gene ablation of zizA causes no change in development, whereas ablation of zizB gives rise to an aberrant developmental morphology and a reduction in cell directionality and velocity, and altered cell shape. Fluorescently labelled ZizA protein associates with the microtubule-organising centre (MTOC), whereas ZizB is enriched in the cortex. Overexpression of ZizB also causes an increase in the number of filopodia and a partial inhibition of cytokinesis. Analysis of ZizB protein binding partners shows that it interacts with Rac1a and a range of actin-associated proteins. In conclusion, our work provides insight into the molecular and cellular functions of zizimin GEF proteins, which are shown to have a role in cell movement, filopodia formation and cytokinesis.


Asunto(s)
Movimiento Celular , Citocinesis , Dictyostelium/crecimiento & desarrollo , Dictyostelium/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas Protozoarias/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Secuencia de Aminoácidos , Dictyostelium/citología , Dictyostelium/genética , Factores de Intercambio de Guanina Nucleótido/química , Factores de Intercambio de Guanina Nucleótido/genética , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Proteínas de Unión al GTP rac/genética
5.
PLoS Biol ; 9(5): e1000618, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21610858

RESUMEN

The mechanism of eukaryotic chemotaxis remains unclear despite intensive study. The most frequently described mechanism acts through attractants causing actin polymerization, in turn leading to pseudopod formation and cell movement. We recently proposed an alternative mechanism, supported by several lines of data, in which pseudopods are made by a self-generated cycle. If chemoattractants are present, they modulate the cycle rather than directly causing actin polymerization. The aim of this work is to test the explanatory and predictive powers of such pseudopod-based models to predict the complex behaviour of cells in chemotaxis. We have now tested the effectiveness of this mechanism using a computational model of cell movement and chemotaxis based on pseudopod autocatalysis. The model reproduces a surprisingly wide range of existing data about cell movement and chemotaxis. It simulates cell polarization and persistence without stimuli and selection of accurate pseudopods when chemoattractant gradients are present. It predicts both bias of pseudopod position in low chemoattractant gradients and--unexpectedly--lateral pseudopod initiation in high gradients. To test the predictive ability of the model, we looked for untested and novel predictions. One prediction from the model is that the angle between successive pseudopods at the front of the cell will increase in proportion to the difference between the cell's direction and the direction of the gradient. We measured the angles between pseudopods in chemotaxing Dictyostelium cells under different conditions and found the results agreed with the model extremely well. Our model and data together suggest that in rapidly moving cells like Dictyostelium and neutrophils an intrinsic pseudopod cycle lies at the heart of cell motility. This implies that the mechanism behind chemotaxis relies on modification of intrinsic pseudopod behaviour, more than generation of new pseudopods or actin polymerization by chemoattractants.


Asunto(s)
Actinas/metabolismo , Quimiotaxis , Dictyostelium/citología , Modelos Teóricos , Seudópodos/fisiología , Polaridad Celular , Dictyostelium/fisiología , Ruido , Polimerizacion , Transfección
6.
J Cell Sci ; 123(Pt 13): 2246-55, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20530573

RESUMEN

Cell division requires the tight coordination of multiple cytoskeletal pathways. The best understood of these involves myosin-II-dependent constriction around the cell equator, but both Dictyostelium and mammalian cells also use a parallel, adhesion-dependent mechanism to generate furrows. We show that the actin nucleation factor SCAR/WAVE is strongly activated during Dictyostelium cytokinesis. This activation localises to large polar protrusions, driving separation of the daughter cells. This continues for 10 minutes after division before the daughter cells revert to normal random motility, indicating that this is a tightly regulated process. We demonstrate that SCAR activity is essential to drive myosin-II-independent cytokinesis, and stabilises the furrow, ensuring symmetrical division. SCAR is also responsible for the generation of MiDASes, mitosis-specific actin-rich adhesions. Loss of SCAR in both Dictyostelium and Drosophila leads to a similar mitotic phenotype, with severe mitotic blebbing, indicating conserved functionality. We also find that the microtubule end-binding protein EB1 is required to restrict SCAR localisation and direct migration. EB1-null cells also exhibit decreased adhesion during mitosis. Our data reveal a spindle-directed signalling pathway that regulates SCAR activity, migration and adhesion at mitosis.


Asunto(s)
Citocinesis/fisiología , Mitosis/fisiología , Miosinas/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Movimiento Celular/fisiología , Dictyostelium/citología , Dictyostelium/fisiología , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Protozoarias/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/fisiología , Huso Acromático/metabolismo
7.
Biochem J ; 436(1): 45-52, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21401524

RESUMEN

I-BAR (inverse-Bin/amphiphysin/Rvs)-domain-containing proteins such as IRSp53 (insulin receptor substrate of 53 kDa) associate with outwardly curved membranes and connect them to proteins involved in actin dynamics. Research on I-BAR proteins has focussed on possible roles in filopod and lamellipod formation, but their full physiological function remains unclear. The social amoeba Dictyostelium encodes a single I-BAR/SH3 (where SH3 is Src homology 3) protein, called IBARa, along with homologues of proteins that interact with IRSp53 family proteins in mammalian cells, providing an excellent model to study its cellular function. Disruption of the gene encoding IBARa leads to a mild defect in development, but filopod and pseudopod dynamics are unaffected. Furthermore, ectopically expressed IBARa does not induce filopod formation and does not localize to filopods. Instead, IBARa associates with clathrin puncta immediately before they are endocytosed. This role is conserved: human BAIAP2L2 (brain-specific angiogenesis inhibitor 1-associated protein 2-like 2) also tightly co-localizes with clathrin plaques, although its homologues IRSp53 and IRTKS (insulin receptor tyrosine kinase substrate) associate with other punctate structures. The results from the present study suggest that I-BAR-containing proteins help generate the membrane curvature required for endocytosis and implies an unexpected role for IRSp53 family proteins in vesicle trafficking.


Asunto(s)
Dictyostelium/metabolismo , Endocitosis/fisiología , Proteínas Protozoarias/metabolismo , Membrana Celular/metabolismo , Clatrina/genética , Clatrina/metabolismo , Células HeLa , Humanos , Proteínas Sustrato del Receptor de Insulina/química , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Dominios Homologos src
8.
Front Immunol ; 13: 1075386, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36524124

RESUMEN

Phagocytosis, macropinocytosis, and G protein coupled receptor-mediated chemotaxis are Ras-regulated and actin-driven processes. The common regulator for Ras activity in these three processes remains unknown. Here, we show that C2GAP2, a Ras GTPase activating protein, highly expressed in the vegetative growth state in model organism Dictyostelium. C2GAP2 localizes at the leading edge of chemotaxing cells, phagosomes during phagocytosis, and macropinosomes during micropinocytosis. c2gapB- cells lacking C2GAP2 displayed increased Ras activation upon folic acid stimulation and subsequent impaired chemotaxis in the folic acid gradient. In addition, c2gaB- cells have elevated phagocytosis and macropinocytosis, which subsequently results in faster cell growth. C2GAP2 binds multiple phospholipids on the plasma membrane and the membrane recruitment of C2GAP2 requires calcium. Taken together, we show a shared negative regulator of Ras signaling that mediates Ras signaling for chemotaxis, phagocytosis, and macropinocytosis.


Asunto(s)
Dictyostelium , Dictyostelium/metabolismo , Quimiotaxis , Pinocitosis/fisiología , Fagocitosis , Ácido Fólico
9.
Plasmid ; 61(2): 119-25, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19046986

RESUMEN

Inducible expression systems are essential for the expression of toxic proteins and are very convenient for proteins that induce strong side effects such as retardation of growth or development. Currently available systems for use in Dictyostelium either do not have a very tight control over expression levels or use a combination of an integrating and an extrachromosomal vector. We designed a new vector in which all components of the available 2-plasmid tetracycline-inducible system were combined onto a single extrachromosomal vector. Two types of inducible plasmids are presented, in which transcription is induced by adding or removing doxycycline, respectively. The location and orientation of the components was optimized in order to obtain a low background expression combined with high inducibility. The resulting vectors have a very low expression in the uninduced state (>1000-fold lower expression compared to that resulting from the act15 promoter), show a 10,000-fold induction of gene expression in a doxycycline concentration-dependent manner and are comparatively small (8.5 kb). With these new vectors, inducible gene expression is as easy as constitutive gene expression.


Asunto(s)
Dictyostelium/genética , Doxiciclina/farmacología , Expresión Génica/efectos de los fármacos , Vectores Genéticos , Plásmidos , Biosíntesis de Proteínas , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Tetraciclina/farmacología
10.
Plasmid ; 61(2): 110-8, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19063918

RESUMEN

A new set of extrachromosomal Dictyostelium expression vectors is presented that can be modified according to the experimental needs with minimal cloning efforts. To achieve this, the vector consists of four functional regions that are separated by unique restriction sites, (1) an Escherichia coli replication region, and regions for (2) replication, (3) selection and (4) protein expression in Dictyostelium. Each region was trimmed down to its smallest possible size. A basic expression vector can be constructed from these modules with a size of only 6.8 kb. By exchanging modules, a large number of vectors with different properties can be constructed. The resulting set of vectors allows most basic expression needs, such as immuno blotting, protein purification, visualization of protein localization and identification of protein-protein interactions. In addition, two genes can be simultaneously expressed on one vector, which yields far more synchronous levels of expression than when expressing two genes on separate plasmids.


Asunto(s)
Clonación Molecular , Dictyostelium/genética , Vectores Genéticos/fisiología , Plásmidos/fisiología , Animales , Escherichia coli/genética , Expresión Génica , Vectores Genéticos/genética , Plásmidos/genética
11.
Mol Biol Cell ; 17(9): 3921-9, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16790492

RESUMEN

Chemotaxis of amoeboid cells is driven by actin filaments in leading pseudopodia and actin-myosin filaments in the back and at the side of the cell to suppress pseudopodia. In Dictyostelium, cGMP plays an important role during chemotaxis and is produced predominantly by a soluble guanylyl cyclase (sGC). The sGC protein is enriched in extending pseudopodia at the leading edge of the cell during chemotaxis. We show here that the sGC protein and the cGMP product have different functions during chemotaxis, using two mutants that lose either catalytic activity (sGCDelta cat) or localization to the leading edge (sGCDeltaN). Cells expressing sGCDeltaN exhibit excellent cGMP formation and myosin localization in the back of the cell, but they exhibit poor orientation at the leading edge. Cells expressing the catalytically dead sGCDelta cat mutant show poor myosin localization at the back, but excellent localization of the sGC protein at the leading edge, where it enhances the probability that a new pseudopod is made in proximity to previous pseudopodia, resulting in a decrease of the degree of turning. Thus cGMP suppresses pseudopod formation in the back of the cell, whereas the sGC protein refines pseudopod formation at the leading edge.


Asunto(s)
Polaridad Celular/fisiología , Quimiotaxis/fisiología , GMP Cíclico/metabolismo , Dictyostelium/citología , Dictyostelium/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Actinas/metabolismo , Animales , Polaridad Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , AMP Cíclico/farmacología , Dictyostelium/efectos de los fármacos , Dictyostelium/enzimología , Guanilato Ciclasa , Proteínas Mutantes/metabolismo , Miosinas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Guanilil Ciclasa Soluble
12.
J Vis Exp ; (143)2019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30735174

RESUMEN

Dictyostelium discoideum is an intriguing model organism for the study of cell differentiation processes during development, cell signaling, and other important cellular biology questions. The technologies available to genetically manipulate Dictyostelium cells are well-developed. Transfections can be performed using different selectable markers and marker re-cycling, including homologous recombination and insertional mutagenesis. This is supported by a well-annotated genome. However, these approaches are optimized for axenic cell lines growing in liquid cultures and are difficult to apply to non-axenic wild-type cells, which feed only on bacteria. The mutations that are present in axenic strains disturb Ras signaling, causing excessive macropinocytosis required for feeding, and impair cell migration, which confounds the interpretation of signal transduction and chemotaxis experiments in those strains. Earlier attempts to genetically manipulate non-axenic cells have lacked efficiency and required complex experimental procedures. We have developed a simple transfection protocol that, for the first time, overcomes these limitations. Those series of large improvements to Dictyostelium molecular genetics allow wild-type cells to be manipulated as easily as standard laboratory strains. In addition to the advantages for studying uncorrupted signaling and motility processes, mutants that disrupt macropinocytosis-based growth can now be readily isolated. Furthermore, the entire transfection workflow is greatly accelerated, with recombinant cells that can be generated in days rather than weeks. Another advantage is that molecular genetics can further be performed with freshly isolated wild-type Dictyostelium samples from the environment. This can help to extend the scope of approaches used in these research areas.


Asunto(s)
Bacterias/crecimiento & desarrollo , Quimiotaxis , Dictyostelium/crecimiento & desarrollo , Ingeniería Genética/métodos , Pinocitosis/fisiología , Bacterias/genética , Dictyostelium/genética , Recombinación Homóloga , Mutagénesis Insercional , Mutación , Transducción de Señal
13.
Sci STKE ; 2007(396): pe40, 2007 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-17652307

RESUMEN

During chemotaxis, phosphatidylinositol 3,4,5-trisphosphate (PIP(3)) accumulates at the leading edge of a eukaryotic cell, where it induces the formation of pseudopodia. PIP(3) has been suggested to be the compass of cells navigating in gradients of signaling molecules. Recent observations suggest that chemotaxis is more complex than previously anticipated. Complete inhibition of all PIP(3) signaling has little effect, and alternative pathways have been identified. In addition, selective pseudopod growth and retraction are more important in directing cell movement than is the place where new pseudopodia are formed.


Asunto(s)
Quimiotaxis/fisiología , Dictyostelium/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Fosfatos de Fosfatidilinositol/fisiología , Fosfolipasas A/fisiología , Proteínas Protozoarias/fisiología , Seudópodos/fisiología , Sistemas de Mensajero Secundario/fisiología , Animales , Factores Quimiotácticos/farmacología , Quimiotaxis/efectos de los fármacos , Cromonas/farmacología , AMP Cíclico/fisiología , GMP Cíclico/fisiología , Citoesqueleto/ultraestructura , Dictyostelium/efectos de los fármacos , Dictyostelium/genética , Dictyostelium/ultraestructura , Relación Dosis-Respuesta a Droga , Guanilato Ciclasa/fisiología , Modelos Biológicos , Morfolinas/farmacología , Concentración Osmolar , Fosfohidrolasa PTEN/fisiología , Fosfatidilinositol 4,5-Difosfato/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosfolipasas A/genética , Fosforilación , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/genética , Procesos Estocásticos
14.
Mol Biol Cell ; 16(2): 976-83, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15601898

RESUMEN

Dictyostelium contains two guanylyl cyclases, GCA, a 12-transmembrane enzyme, and sGC, a homologue of mammalian soluble adenylyl cyclase. sGC provides nearly all chemoattractant-stimulated cGMP formation and is essential for efficient chemotaxis toward cAMP. We show that in resting cells the major fraction of the sGC-GFP fusion protein localizes to the cytosol, and a small fraction is associated to the cell cortex. With the artificial substrate Mn2+/GTP, sGC activity and protein exhibit a similar distribution between soluble and particulate fraction of cell lysates. However, with the physiological substrate Mg2+/GTP, sGC in the cytosol is nearly inactive, whereas the particulate enzyme shows high enzyme activity. Reconstitution experiments reveal that inactive cytosolic sGC acquires catalytic activity with Mg2+/GTP upon association to the membrane. Stimulation of cells with cAMP results in a twofold increase of membrane-localized sGC-GFP, which is accompanied by an increase of the membrane-associated guanylyl cyclase activity. In a cAMP gradient, sGC-GFP localizes to the anterior cell cortex, suggesting that in chemotacting cells, sGC is activated at the leading edge of the cell.


Asunto(s)
Membrana Celular/metabolismo , Quimiotaxis , Dictyostelium/enzimología , Guanilato Ciclasa/metabolismo , Animales , Catálisis , Polaridad Celular , AMP Cíclico/farmacología , GMP Cíclico/biosíntesis , Citosol/metabolismo , Dictyostelium/genética , Activación Enzimática , Proteínas Fluorescentes Verdes/metabolismo , Guanosina Trifosfato/metabolismo , Guanilato Ciclasa/química , Guanilato Ciclasa/efectos de los fármacos , Guanilato Ciclasa/genética , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Magnesio/farmacología , Manganeso/farmacología , Fusión de Membrana , Solubilidad , Especificidad por Sustrato
15.
PLoS One ; 13(5): e0196809, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29847546

RESUMEN

Dictyostelium has a mature technology for molecular-genetic manipulation based around transfection using several different selectable markers, marker re-cycling, homologous recombination and insertional mutagenesis, all supported by a well-annotated genome. However this technology is optimized for mutant, axenic cells that, unlike non-axenic wild type, can grow in liquid medium. There is a pressing need for methods to manipulate wild type cells and ones with defects in macropinocytosis, neither of which can grow in liquid media. Here we present a panel of molecular genetic techniques based on the selection of Dictyostelium transfectants by growth on bacteria rather than liquid media. As well as extending the range of strains that can be manipulated, these techniques are faster than conventional methods, often giving usable numbers of transfected cells within a few days. The methods and plasmids described here allow efficient transfection with extrachromosomal vectors, as well as chromosomal integration at a 'safe haven' for relatively uniform cell-to-cell expression, efficient gene knock-in and knock-out and an inducible expression system. We have thus created a complete new system for the genetic manipulation of Dictyostelium cells that no longer requires cell feeding on liquid media.


Asunto(s)
Dictyostelium/genética , Técnicas de Sustitución del Gen/métodos , Ingeniería Genética/métodos , Vectores Genéticos/genética , Recombinación Homóloga/genética , Mutagénesis Insercional/genética , Mutación/genética , Pinocitosis/genética , Plásmidos/genética , Transfección/métodos
16.
Elife ; 52016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27960076

RESUMEN

Macropinocytosis is a fundamental mechanism that allows cells to take up extracellular liquid into large vesicles. It critically depends on the formation of a ring of protrusive actin beneath the plasma membrane, which develops into the macropinocytic cup. We show that macropinocytic cups in Dictyostelium are organised around coincident intense patches of PIP3, active Ras and active Rac. These signalling patches are invariably associated with a ring of active SCAR/WAVE at their periphery, as are all examined structures based on PIP3 patches, including phagocytic cups and basal waves. Patch formation does not depend on the enclosing F-actin ring, and patches become enlarged when the RasGAP NF1 is mutated, showing that Ras plays an instructive role. New macropinocytic cups predominantly form by splitting from existing ones. We propose that cup-shaped plasma membrane structures form from self-organizing patches of active Ras/PIP3, which recruit a ring of actin nucleators to their periphery.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Membrana Celular/metabolismo , Dictyostelium/fisiología , Pinocitosis , Fosfatidilinositoles/metabolismo , Proteínas Protozoarias/metabolismo , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteínas ras/metabolismo
17.
Dev Cell ; 37(5): 458-72, 2016 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-27237792

RESUMEN

Chemotaxis, or directional movement toward extracellular chemical gradients, is an important property of cells that is mediated through G-protein-coupled receptors (GPCRs). Although many chemotaxis pathways downstream of Gßγ have been identified, few Gα effectors are known. Gα effectors are of particular importance because they allow the cell to distinguish signals downstream of distinct chemoattractant GPCRs. Here we identify GflB, a Gα2 binding partner that directly couples the Dictyostelium cyclic AMP GPCR to Rap1. GflB localizes to the leading edge and functions as a Gα-stimulated, Rap1-specific guanine nucleotide exchange factor required to balance Ras and Rap signaling. The kinetics of GflB translocation are fine-tuned by GSK-3 phosphorylation. Cells lacking GflB display impaired Rap1/Ras signaling and actin and myosin dynamics, resulting in defective chemotaxis. Our observations demonstrate that GflB is an essential upstream regulator of chemoattractant-mediated cell polarity and cytoskeletal reorganization functioning to directly link Gα activation to monomeric G-protein signaling.


Asunto(s)
Quimiotaxis , Dictyostelium/citología , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas Protozoarias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Actinas/metabolismo , Quimiotaxis/efectos de los fármacos , AMP Cíclico/farmacología , Dictyostelium/efectos de los fármacos , Dictyostelium/metabolismo , Activación Enzimática/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Modelos Biológicos , Miosina Tipo II/metabolismo , Fosforilación/efectos de los fármacos , Polimerizacion/efectos de los fármacos , Proteínas ras/metabolismo
18.
Biochim Biophys Acta ; 1623(2-3): 129-34, 2003 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-14572910

RESUMEN

Chemoattractant stimulation of Dictyostelium cells leads to the opening of calcium channels in the plasma membrane, causing extracellular calcium to flux into the cell. The genetically uncharacterised mutants stmF and KI8 show strongly altered chemoattractant-stimulated cGMP responses. The aberrant calcium influx in these strains has provided evidence that the chemoattractant-stimulated calcium influx is potentiated by cGMP. We have tested this hypothesis in genetically defined mutants by measuring the calcium influx in a strain that lacks intracellular cGMP due to the disruption of two guanylyl cyclases, and in a strain with increased cGMP levels caused by the disruption of two cGMP-degrading phosphodiesterases. The results reveal that the calcium influx stimulated by cAMP or folic acid is essentially identical in these strains. We conclude that cGMP is not involved in chemoattractant-stimulated calcium influx.


Asunto(s)
Señalización del Calcio/fisiología , GMP Cíclico/metabolismo , Dictyostelium/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , AMP Cíclico/farmacología , Dictyostelium/efectos de los fármacos , Dictyostelium/genética , Ácido Fólico/farmacología , Genes Protozoarios , Cinética , Mutación
19.
Elife ; 42015 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-25815683

RESUMEN

Cells use phagocytosis and macropinocytosis to internalise bulk material, which in phagotrophic organisms supplies the nutrients necessary for growth. Wildtype Dictyostelium amoebae feed on bacteria, but for decades laboratory work has relied on axenic mutants that can also grow on liquid media. We used forward genetics to identify the causative gene underlying this phenotype. This gene encodes the RasGAP Neurofibromin (NF1). Loss of NF1 enables axenic growth by increasing fluid uptake. Mutants form outsized macropinosomes which are promoted by greater Ras and PI3K activity at sites of endocytosis. Relatedly, NF1 mutants can ingest larger-than-normal particles using phagocytosis. An NF1 reporter is recruited to nascent macropinosomes, suggesting that NF1 limits their size by locally inhibiting Ras signalling. Our results link NF1 with macropinocytosis and phagocytosis for the first time, and we propose that NF1 evolved in early phagotrophs to spatially modulate Ras activity, thereby constraining and shaping their feeding structures.


Asunto(s)
Dictyostelium/genética , Neurofibromina 1/genética , Fagocitosis/genética , Pinocitosis/genética , Proteínas Protozoarias/genética , Gránulos Citoplasmáticos/genética , Gránulos Citoplasmáticos/metabolismo , Dictyostelium/crecimiento & desarrollo , Dictyostelium/metabolismo , Endocitosis/genética , Mutación , Neurofibromina 1/metabolismo , Fagosomas/genética , Fagosomas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Protozoarias/metabolismo , Transducción de Señal/genética , Proteínas ras/metabolismo
20.
J Cell Biol ; 204(4): 497-505, 2014 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-24535823

RESUMEN

In eukaryotic chemotaxis, the mechanisms connecting external signals to the motile apparatus remain unclear. The role of the lipid phosphatidylinositol 3,4,5-trisphosphate (PIP3) has been particularly controversial. PIP3 has many cellular roles, notably in growth control and macropinocytosis as well as cell motility. Here we show that PIP3 is not only unnecessary for Dictyostelium discoideum to migrate toward folate, but actively inhibits chemotaxis. We find that macropinosomes, but not pseudopods, in growing cells are dependent on PIP3. PIP3 patches in these cells show no directional bias, and overall only PIP3-free pseudopods orient up-gradient. The pseudopod driver suppressor of cAR mutations (SCAR)/WASP and verprolin homologue (WAVE) is not recruited to the center of PIP3 patches, just the edges, where it causes macropinosome formation. Wild-type cells, unlike the widely used axenic mutants, show little macropinocytosis and few large PIP3 patches, but migrate more efficiently toward folate. Tellingly, folate chemotaxis in axenic cells is rescued by knocking out phosphatidylinositide 3-kinases (PI 3-kinases). Thus PIP3 promotes macropinocytosis and interferes with pseudopod orientation during chemotaxis of growing cells.


Asunto(s)
Quimiotaxis/fisiología , Dictyostelium/fisiología , Ácido Fólico/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Pinocitosis/fisiología , Movimiento Celular/fisiología , Mutación/genética , Proteínas Protozoarias/genética , Seudópodos , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA