Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 626(7999): 635-642, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38297127

RESUMEN

Type 2 diabetes mellitus is a major risk factor for hepatocellular carcinoma (HCC). Changes in extracellular matrix (ECM) mechanics contribute to cancer development1,2, and increased stiffness is known to promote HCC progression in cirrhotic conditions3,4. Type 2 diabetes mellitus is characterized by an accumulation of advanced glycation end-products (AGEs) in the ECM; however, how this affects HCC in non-cirrhotic conditions is unclear. Here we find that, in patients and animal models, AGEs promote changes in collagen architecture and enhance ECM viscoelasticity, with greater viscous dissipation and faster stress relaxation, but not changes in stiffness. High AGEs and viscoelasticity combined with oncogenic ß-catenin signalling promote HCC induction, whereas inhibiting AGE production, reconstituting the AGE clearance receptor AGER1 or breaking AGE-mediated collagen cross-links reduces viscoelasticity and HCC growth. Matrix analysis and computational modelling demonstrate that lower interconnectivity of AGE-bundled collagen matrix, marked by shorter fibre length and greater heterogeneity, enhances viscoelasticity. Mechanistically, animal studies and 3D cell cultures show that enhanced viscoelasticity promotes HCC cell proliferation and invasion through an integrin-ß1-tensin-1-YAP mechanotransductive pathway. These results reveal that AGE-mediated structural changes enhance ECM viscoelasticity, and that viscoelasticity can promote cancer progression in vivo, independent of stiffness.


Asunto(s)
Carcinoma Hepatocelular , Progresión de la Enfermedad , Elasticidad , Matriz Extracelular , Cirrosis Hepática , Neoplasias Hepáticas , Animales , Humanos , beta Catenina/metabolismo , Carcinoma Hepatocelular/complicaciones , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proliferación Celular , Colágeno/química , Colágeno/metabolismo , Simulación por Computador , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Matriz Extracelular/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Integrina beta1/metabolismo , Neoplasias Hepáticas/complicaciones , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Invasividad Neoplásica , Viscosidad , Proteínas Señalizadoras YAP/metabolismo , Cirrosis Hepática/complicaciones , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología
2.
Nat Rev Mol Cell Biol ; 14(7): 405-15, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23778968

RESUMEN

Recently, a consensus has emerged that cofilin severing activity can generate free actin filament ends that are accessible for F-actin polymerization and depolymerization without changing the rate of G-actin association and dissociation at either filament end. The structural basis of actin filament severing by cofilin is now better understood. These results have been integrated with recently discovered mechanisms for cofilin activation in migrating cells, which led to new models for cofilin function that provide insights into how cofilin regulation determines the temporal and spatial control of cell behaviour.


Asunto(s)
Factores Despolimerizantes de la Actina/fisiología , Movimiento Celular , Factores Despolimerizantes de la Actina/química , Actinas/metabolismo , Animales , Extensiones de la Superficie Celular/metabolismo , Humanos , Modelos Moleculares , Fosforilación , Multimerización de Proteína , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Transporte de Proteínas
3.
Nat Mater ; 2023 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-37957268

RESUMEN

Breast cancer becomes invasive when carcinoma cells invade through the basement membrane (BM)-a nanoporous layer of matrix that physically separates the primary tumour from the stroma. Single cells can invade through nanoporous three-dimensional matrices due to protease-mediated degradation or force-mediated widening of pores via invadopodial protrusions. However, how multiple cells collectively invade through the physiological BM, as they do during breast cancer progression, remains unclear. Here we developed a three-dimensional in vitro model of collective invasion of the BM during breast cancer. We show that cells utilize both proteases and forces-but not invadopodia-to breach the BM. Forces are generated from a combination of global cell volume expansion, which stretches the BM, and local contractile forces that act in the plane of the BM to breach it, allowing invasion. These results uncover a mechanism by which cells collectively interact to overcome a critical barrier to metastasis.

4.
J Microsc ; 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38357769

RESUMEN

Genetically encoded, fluorescent protein (FP)-based Förster resonance energy transfer (FRET) biosensors are microscopy imaging tools tailored for the precise monitoring and detection of molecular dynamics within subcellular microenvironments. They are characterised by their ability to provide an outstanding combination of spatial and temporal resolutions in live-cell microscopy. In this review, we begin by tracing back on the historical development of genetically encoded FP labelling for detection in live cells, which lead us to the development of early biosensors and finally to the engineering of single-chain FRET-based biosensors that have become the state-of-the-art today. Ultimately, this review delves into the fundamental principles of FRET and the design strategies underpinning FRET-based biosensors, discusses their diverse applications and addresses the distinct challenges associated with their implementation. We place particular emphasis on single-chain FRET biosensors for the Rho family of guanosine triphosphate hydrolases (GTPases), pointing to their historical role in driving our understanding of the molecular dynamics of this important class of signalling proteins and revealing the intricate relationships and regulatory mechanisms that comprise Rho GTPase biology in living cells.

5.
Exp Cell Res ; 433(2): 113852, 2023 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-37951335

RESUMEN

In the study of tumorigenesis, the involvement of molecules within the extracellular matrix (ECM) is crucial. ADAMTSs (A Disintegrin and Metalloproteinase with Thrombospondin motifs), a group of secreted proteases known for their role in ECM remodeling, were primarily considered to be extracellular proteases. However, our research specifically detected ADAMTS-1, a member of this family, predominantly within the nucleus of mammary cells. Our main objective was to understand the mechanism of ADAMTS-1 translocation to the nucleus and its functional significance in this cellular compartment. Our investigation uncovered that nuclear ADAMTS-1 was present in cells exhibiting an epithelial phenotype, while cells of mesenchymal origin contained the protease in the cytoplasm. Moreover, disruption of ADAMTS-1 secretion, induced by Monensin treatment, resulted in its accumulation in the cytoplasm. Notably, our research indicated that alterations in the secretory pathways could influence the protease's compartmentalization. Additionally, experiments with conditioned medium from cells containing nuclear ADAMTS-1 demonstrated its internalization into the nucleus by HT-1080 cells and fibroblasts. Furthermore, heightened levels of ADAMTS-1 within the ECM reduced the migratory potential of mesenchymal cells. This highlights the potential significance of nuclear ADAMTS-1 as a critical component within the tumor microenvironment due to its functional activity in this specific cellular compartment.


Asunto(s)
Proteína ADAMTS1 , Movimiento Celular , Núcleo Celular , Matriz Extracelular , Trombospondinas , Humanos , Proteína ADAMTS1/genética , Proteína ADAMTS1/metabolismo , Carcinogénesis/metabolismo , Endopeptidasas/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Trombospondinas/metabolismo , Microambiente Tumoral , Núcleo Celular/metabolismo
6.
Biophys J ; 122(18): 3600-3610, 2023 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-36523161

RESUMEN

The microtubule (MT) cytoskeleton and its dynamics play an important role in cell migration. Depletion of the microtubule-severing enzyme Fidgetin-like 2 (FL2), a regulator of MT dynamics at the leading edge of migrating cells, leads to faster and more efficient cell migration. Here we examine how siRNA knockdown of FL2 increases cell motility. Förster resonance energy transfer biosensor studies shows that FL2 knockdown decreases activation of the p21 Rho GTPase, RhoA, and its activator GEF-H1. Immunofluorescence studies reveal that GEF-H1 is sequestered by the increased MT density resulting from FL2 depletion. Activation of the Rho GTPase, Rac1, however, does not change after FL2 knockdown. Furthermore, FL2 depletion leads to an increase in focal adhesion kinase activation at the leading edge, as shown by immunofluorescence studies, but no change in actin dynamics, as shown by fluorescence recovery after photobleaching. We believe these results expand our understanding of the role of MT dynamics in cell migration and offer new insights into RhoA and Rac1 regulation.


Asunto(s)
Microtúbulos , Proteína de Unión al GTP rhoA , Factores de Intercambio de Guanina Nucleótido Rho/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Microtúbulos/metabolismo , Movimiento Celular , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo , Actinas/metabolismo , Proteínas de Unión al GTP rho/metabolismo
7.
Exp Cell Res ; 410(2): 112939, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34813733

RESUMEN

One of the hallmarks of cancer cells is their exceptional ability to migrate within the extracellular matrix (ECM) for gaining access to the circulatory system, a critical step of cancer metastasis. RhoA, a small GTPase, is known to be a key molecular switch that toggles between actomyosin contractility and lamellipodial protrusion during cell migration. Current understanding of RhoA activity in cell migration has been largely derived from studies of cells plated on a two-dimensional (2D) substrate using a FRET biosensor. There has been increasing evidence that cells behave differently in a more physiologically relevant three-dimensional (3D) environment. However, studies of RhoA activities in 3D have been hindered by low signal-to-noise ratio in fluorescence imaging. In this paper, we present a a machine learning-assisted FRET technique to follow the spatiotemporal dynamics of RhoA activities of single breast tumor cells (MDA-MB-231) migrating in a 3D as well as a 2D environment. We found that RhoA activity is more polarized along the long axis of the cell for single cells migrating on 2D fibronectin-coated glass versus those embedded in 3D collagen matrices. In particular, RhoA activities of cells in 2D exhibit a distinct front-to-back and back-to-front movement during migration in contrast to those in 3D. Finally, regardless of dimensionality, RhoA polarization is found to be moderately correlated with cell shape.


Asunto(s)
Neoplasias de la Mama/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Aprendizaje Automático , Proteína de Unión al GTP rhoA/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular , Polaridad Celular , Forma de la Célula , Colágeno/metabolismo , Femenino , Humanos , Ratas , Factores de Tiempo
8.
Genes Dev ; 29(8): 876-86, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25877922

RESUMEN

Repetitive nucleotide or amino acid sequences are often engineered into probes and biosensors to achieve functional readouts and robust signal amplification. However, these repeated sequences are notoriously prone to aberrant deletion and degradation, impacting the ability to correctly detect and interpret biological functions. Here, we introduce a facile and generalizable approach to solve this often unappreciated problem by modifying the nucleotide sequences of the target mRNA to make them nonrepetitive but still functional ("synonymous"). We first demonstrated the procedure by designing a cassette of synonymous MS2 RNA motifs and tandem coat proteins for RNA imaging and showed a dramatic improvement in signal and reproducibility in single-RNA detection in live cells. The same approach was extended to enhancing the stability of engineered fluorescent biosensors containing a fluorescent resonance energy transfer (FRET) pair of fluorescent proteins on which a great majority of systems thus far in the field are based. Using the synonymous modification to FRET biosensors, we achieved correct expression of full-length sensors, eliminating the aberrant truncation products that often were assumed to be due to nonspecific proteolytic cleavages. Importantly, the biological interpretations of the sensor are significantly different when a correct, full-length biosensor is expressed. Thus, we show here a useful and generally applicable method to maintain the integrity of expressed genes, critical for the correct interpretation of probe readouts.


Asunto(s)
Expresión Génica , Técnicas Genéticas , Secuencias Repetitivas de Ácidos Nucleicos/genética , Animales , Secuencia de Bases/genética , Proteínas de la Cápside/genética , Línea Celular , Células Cultivadas , Codón/genética , Humanos , Levivirus/genética , Ratones , Motivos de Nucleótidos , Saccharomyces cerevisiae/genética
9.
Nature ; 539(7630): 575-578, 2016 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-27828948

RESUMEN

Mitochondrial products such as ATP, reactive oxygen species, and aspartate are key regulators of cellular metabolism and growth. Abnormal mitochondrial function compromises integrated growth-related processes such as development and tissue repair, as well as homeostatic mechanisms that counteract ageing and neurodegeneration, cardiovascular disease, and cancer. Physiologic mechanisms that control mitochondrial activity in such settings remain incompletely understood. Here we show that the atypical Fat1 cadherin acts as a molecular 'brake' on mitochondrial respiration that regulates vascular smooth muscle cell (SMC) proliferation after arterial injury. Fragments of Fat1 accumulate in SMC mitochondria, and the Fat1 intracellular domain interacts with multiple mitochondrial proteins, including critical factors associated with the inner mitochondrial membrane. SMCs lacking Fat1 (Fat1KO) grow faster, consume more oxygen for ATP production, and contain more aspartate. Notably, expression in Fat1KO cells of a modified Fat1 intracellular domain that localizes exclusively to mitochondria largely normalizes oxygen consumption, and the growth advantage of these cells can be suppressed by inhibition of mitochondrial respiration, which suggest that a Fat1-mediated growth control mechanism is intrinsic to mitochondria. Consistent with this idea, Fat1 species associate with multiple respiratory complexes, and Fat1 deletion both increases the activity of complexes I and II and promotes the formation of complex-I-containing supercomplexes. In vivo, Fat1 is expressed in injured human and mouse arteries, and inactivation of SMC Fat1 in mice potentiates the response to vascular damage, with markedly increased medial hyperplasia and neointimal growth, and evidence of higher SMC mitochondrial respiration. These studies suggest that Fat1 controls mitochondrial activity to restrain cell growth during the reparative, proliferative state induced by vascular injury. Given recent reports linking Fat1 to cancer, abnormal kidney and muscle development, and neuropsychiatric disease, this Fat1 function may have importance in other settings of altered cell growth and metabolism.


Asunto(s)
Arterias/citología , Arterias/metabolismo , Cadherinas/metabolismo , Respiración de la Célula , Mitocondrias/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Aorta/citología , Aorta/lesiones , Aorta/metabolismo , Arterias/lesiones , Ácido Aspártico/metabolismo , Cadherinas/química , Cadherinas/deficiencia , Proliferación Celular , Técnicas de Inactivación de Genes , Humanos , Masculino , Ratones , Mitocondrias/química , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/metabolismo , Neointima/metabolismo , Oxígeno/metabolismo , Consumo de Oxígeno
10.
Nature ; 540(7631): 74-79, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27775718

RESUMEN

Mitochondria are dynamic organelles that exchange contents and undergo remodelling during cyclic fusion and fission. Genetic mutations in MFN2 (the gene encoding mitofusin 2) interrupt mitochondrial fusion and cause the untreatable neurodegenerative condition Charcot-Marie-Tooth disease type 2A (CMT2A). It has not yet been possible to directly modulate mitochondrial fusion, in part because the structural basis of mitofusin function is not completely understood. Here we show that mitofusins adopt either a fusion-constrained or a fusion-permissive molecular conformation, directed by specific intramolecular binding interactions, and demonstrate that mitofusin-dependent mitochondrial fusion can be regulated in mouse cells by targeting these conformational transitions. On the basis of this model, we engineered a cell-permeant minipeptide to destabilize the fusion-constrained conformation of mitofusin and promote the fusion-permissive conformation, reversing mitochondrial abnormalities in cultured fibroblasts and neurons that harbour CMT2A-associated genetic defects. The relationship between the conformational plasticity of mitofusin 2 and mitochondrial dynamism reveals a central mechanism that regulates mitochondrial fusion, the manipulation of which can correct mitochondrial pathology triggered by defective or imbalanced mitochondrial dynamics.


Asunto(s)
GTP Fosfohidrolasas/química , GTP Fosfohidrolasas/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Dinámicas Mitocondriales/efectos de los fármacos , Péptidos/farmacología , Animales , Células Cultivadas , Enfermedad de Charcot-Marie-Tooth/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , GTP Fosfohidrolasas/genética , Ratones , Mitocondrias/genética , Mitocondrias/patología , Dinámicas Mitocondriales/genética , Modelos Moleculares , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Péptidos/química , Permeabilidad , Conformación Proteica/efectos de los fármacos
11.
Nat Chem Biol ; 14(6): 591-600, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29686359

RESUMEN

Direct visualization and light control of several cellular processes is a challenge, owing to the spectral overlap of available genetically encoded probes. Here we report the most red-shifted monomeric near-infrared (NIR) fluorescent protein, miRFP720, and the fully NIR Förster resonance energy transfer (FRET) pair miRFP670-miRFP720, which together enabled design of biosensors compatible with CFP-YFP imaging and blue-green optogenetic tools. We developed a NIR biosensor for Rac1 GTPase and demonstrated its use in multiplexed imaging and light control of Rho GTPase signaling pathways. Specifically, we combined the Rac1 biosensor with CFP-YFP FRET biosensors for RhoA and for Rac1-GDI binding, and concurrently used the LOV-TRAP tool for upstream Rac1 activation. We directly observed and quantified antagonism between RhoA and Rac1 dependent on the RhoA-downstream effector ROCK; showed that Rac1 activity and GDI binding closely depend on the spatiotemporal coordination between these two molecules; and simultaneously observed Rac1 activity during optogenetic manipulation of Rac1.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia , Proteínas de Unión al GTP rho/química , Animales , Técnicas Biosensibles , Bradyrhizobium , Células HEK293 , Células HeLa , Humanos , Ratones , Células 3T3 NIH , Optogenética , Plásmidos , Unión Proteica , Transducción de Señal , Espectroscopía Infrarroja Corta , Proteína de Unión al GTP cdc42/química , Proteína de Unión al GTP rac1/química
12.
Nat Chem Biol ; 14(9): 902, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29728601

RESUMEN

In the version of this article originally published, the values for time shown on the x axis of Figure 5c were incorrect. The error has been corrected in all versions of the paper.

13.
Cell Commun Signal ; 18(1): 144, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32900380

RESUMEN

BACKGROUND: Lung cancer is the second most commonly occurring cancer. The ability to metastasize and spread to distant locations renders the tumor more aggressive. Members of the Rho subfamily of small GTP-binding proteins (GTPases) play a central role in the regulation of the actin cytoskeleton and in cancer cell migration and metastasis. In this study we investigated the role of the RhoA/Cdc42 GAP, StarD13, a previously described tumor suppressor, in malignancy, migration and invasion of the lung cancer cells A549. METHODS: We knocked down StarD13 expression in A549 lung cancer cells and tested the effect on cell migration and invadopodia formation using time lapse imaging and invasion assays. We also performed rescue experiments to determine the signaling pathways downstream of StarD13 and transfected the cells with FRET biosensors for RhoGTPases to identify the proteins involved in invadopodia formation. RESULTS: We observed a decrease in the level of expression of StarD13 in lung tumor tissues compared to normal lung tissues through immunohistochemistry. StarD13 also showed a lower expression in the lung adenocarcinoma cell line A549 compared to normal lung cells, WI38. In addition, the depletion of StarD13 increased cell proliferation and viability in WI38 and A549 cells, suggesting that StarD13 might potentially be a tumor suppressor in lung cancer. The depletion of StarD13, however, inhibited cell motility, conversely demonstrating a positive regulatory role in cell migration. This was potentially due to the constitutive activation of RhoA detected by pull down and FRET assays. Surprisingly, StarD13 suppressed cell invasion by inhibiting Cdc42-mediated invadopodia formation. Indeed, TKS4 staining and invadopodia assay revealed that StarD13 depletion increased Cdc42 activation as well as invadopodia formation and matrix degradation. Normal lung cells depleted of StarD13 also produced invadopodia, otherwise a unique hallmark of invasive cancer cells. Cdc42 knock down mimicked the effects of StarD13, while overexpression of a constitutively active Cdc42 mimicked the effects of its depletion. Finally, immunostaining and FRET analysis revealed the absence of StarD13 in invadopodia as compared to Cdc42, which was activated in invadopodia at the sites of matrix degradation. CONCLUSION: In conclusion, StarD13 plays distinct roles in lung cancer cell migration and invasion through its differential regulation of Rho GTPases. Video abstract.


Asunto(s)
Adenocarcinoma del Pulmón/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Neoplasias Pulmonares/metabolismo , Podosomas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Células A549 , Adenocarcinoma del Pulmón/patología , Movimiento Celular , Humanos , Neoplasias Pulmonares/patología , Invasividad Neoplásica/patología , Podosomas/patología
14.
Nat Chem Biol ; 12(10): 802-809, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27501396

RESUMEN

Guanine-nucleotide dissociation inhibitors (GDIs) are negative regulators of Rho family GTPases that sequester the GTPases away from the membrane. Here we ask how GDI-Cdc42 interaction regulates localized Cdc42 activation for cell motility. The sensitivity of cells to overexpression of Rho family pathway components led us to a new biosensor, GDI.Cdc42 FLARE, in which Cdc42 is modified with a fluorescence resonance energy transfer (FRET) 'binding antenna' that selectively reports Cdc42 binding to endogenous GDIs. Similar antennae could also report GDI-Rac1 and GDI-RhoA interaction. Through computational multiplexing and simultaneous imaging, we determined the spatiotemporal dynamics of GDI-Cdc42 interaction and Cdc42 activation during cell protrusion and retraction. This revealed remarkably tight coordination of GTPase release and activation on a time scale of 10 s, suggesting that GDI-Cdc42 interactions are a critical component of the spatiotemporal regulation of Cdc42 activity, and not merely a mechanism for global sequestration of an inactivated pool of signaling molecules.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia , Inhibidores de Disociación de Guanina Nucleótido/química , Inhibidores de Disociación de Guanina Nucleótido/metabolismo , Proteína de Unión al GTP cdc42/química , Proteína de Unión al GTP cdc42/metabolismo , Sitios de Unión , Células HEK293 , Humanos , Análisis Espacio-Temporal
15.
J Immunol ; 196(8): 3479-93, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26951800

RESUMEN

Despite the 92% homology of the hematopoietic cell-specific Rac2 to the canonical isoform Rac1, these isoforms have been shown to play nonredundant roles in immune cells. To study isoform-specific dynamics of Rac in live cells, we developed a genetically encoded, single-chain FRET-based biosensor for Rac2. We also made significant improvements to our existing single-chain Rac1 biosensor. We optimized the biosensor constructs for facile expression in hematopoietic cells and performed functional validations in murine macrophage sublines of RAW264.7 cells. Rac2, Rac1, and Cdc42 have been implicated in the formation of actin-rich protrusions by macrophages, but their individual activation dynamics have not been previously characterized. We found that both Rac1 and Rac2 had similar activation kinetics, yet they had distinct spatial distributions in response to the exogenous stimulus, fMLF. Active Rac1 was mainly localized to the cell periphery, whereas active Rac2 was distributed throughout the cell, with an apparent higher concentration in the perinuclear region. We also performed an extensive morphodynamic analysis of Rac1, Rac2, and Cdc42 activities during the extension of random protrusions. We found that Rac2 appears to play a leading role in the generation of random protrusions, as we observed an initial strong activation of Rac2 in regions distal from the leading edge, followed by the activation of Rac1, a second burst of Rac2 and then Cdc42 immediately behind the leading edge. Overall, isoform-specific biosensors that have been optimized for expression should be valuable for interrogating the coordination of Rho family GTPase activities in living cells.


Asunto(s)
Técnicas Biosensibles/métodos , Transferencia Resonante de Energía de Fluorescencia/métodos , Neuropéptidos/genética , Isoformas de Proteínas/genética , Proteína de Unión al GTP cdc42/genética , Proteínas de Unión al GTP rac/genética , Proteína de Unión al GTP rac1/genética , Animales , Línea Celular , Extensiones de la Superficie Celular/fisiología , Células HEK293 , Humanos , Macrófagos/inmunología , Ratones , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteína RCA2 de Unión a GTP
16.
J Cell Sci ; 126(Pt 15): 3356-69, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23704350

RESUMEN

Protrusion formation is the first step that precedes cell movement of motile cells. Spatial control of actin polymerization is necessary to achieve directional protrusion during cell migration. Here we show that the spatial coordinators p190RhoGEF and p190RhoGAP regulate actin polymerization during leading edge protrusions by regulating the actin barbed end distribution and amplitude. The distribution of RhoC activity and proper balance of cofilin activation achieved by p190RhoGEF and p190RhoGAP determines the direction of final protrusive activity. These findings provide a new insight into the dynamic plasticity in the amplitude and distribution of barbed ends, which can be modulated by fine-tuning RhoC activity by upstream GEFs and GAPs for directed cell motility.


Asunto(s)
Citoesqueleto de Actina/ultraestructura , Actinas/metabolismo , Actinas/ultraestructura , Proteínas de Unión al GTP rho/metabolismo , Citoesqueleto de Actina/química , Citoesqueleto de Actina/patología , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Quimiotaxis/fisiología , Neoplasias Mamarias Experimentales/patología , Ratas , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transfección , Proteínas de Unión al GTP rho/química , Proteínas de Unión al GTP rho/genética
17.
Nature ; 461(7260): 99-103, 2009 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-19693013

RESUMEN

The GTPases Rac1, RhoA and Cdc42 act together to control cytoskeleton dynamics. Recent biosensor studies have shown that all three GTPases are activated at the front of migrating cells, and biochemical evidence suggests that they may regulate one another: Cdc42 can activate Rac1 (ref. 8), and Rac1 and RhoA are mutually inhibitory. However, their spatiotemporal coordination, at the seconds and single-micrometre dimensions typical of individual protrusion events, remains unknown. Here we examine GTPase coordination in mouse embryonic fibroblasts both through simultaneous visualization of two GTPase biosensors and using a 'computational multiplexing' approach capable of defining the relationships between multiple protein activities visualized in separate experiments. We found that RhoA is activated at the cell edge synchronous with edge advancement, whereas Cdc42 and Rac1 are activated 2 micro-m behind the edge with a delay of 40 s. This indicates that Rac1 and RhoA operate antagonistically through spatial separation and precise timing, and that RhoA has a role in the initial events of protrusion, whereas Rac1 and Cdc42 activate pathways implicated in reinforcement and stabilization of newly expanded protrusions.


Asunto(s)
Extensiones de la Superficie Celular/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Técnicas Biosensibles , Movimiento Celular , Forma de la Célula , Embrión de Mamíferos/citología , Activación Enzimática , Fibroblastos/citología , Fibroblastos/enzimología , Ratones , Neuropéptidos/metabolismo , Transporte de Proteínas , Factores de Tiempo , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1 , Proteína de Unión al GTP rhoA
18.
Nat Commun ; 14(1): 8499, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38129387

RESUMEN

Heterozygous deletions in the ANKS1B gene cause ANKS1B neurodevelopmental syndrome (ANDS), a rare genetic disease characterized by autism spectrum disorder (ASD), attention deficit/hyperactivity disorder, and speech and motor deficits. The ANKS1B gene encodes for AIDA-1, a protein that is enriched at neuronal synapses and regulates synaptic plasticity. Here we report an unexpected role for oligodendroglial deficits in ANDS pathophysiology. We show that Anks1b-deficient mouse models display deficits in oligodendrocyte maturation, myelination, and Rac1 function, and recapitulate white matter abnormalities observed in ANDS patients. Selective loss of Anks1b from the oligodendrocyte lineage, but not from neuronal populations, leads to deficits in social preference and sensory reactivity previously observed in a brain-wide Anks1b haploinsufficiency model. Furthermore, we find that clemastine, an antihistamine shown to increase oligodendrocyte precursor cell maturation and central nervous system myelination, rescues deficits in social preference in 7-month-old Anks1b-deficient mice. Our work shows that deficits in social behaviors present in ANDS may originate from abnormal Rac1 activity within oligodendrocytes.


Asunto(s)
Trastorno del Espectro Autista , Animales , Humanos , Lactante , Ratones , Trastorno del Espectro Autista/genética , Péptidos y Proteínas de Señalización Intracelular , Neuronas , Oligodendroglía , Conducta Social
19.
Nature ; 440(7087): 1069-72, 2006 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-16547516

RESUMEN

Rho family GTPases regulate the actin and adhesion dynamics that control cell migration. Current models postulate that Rac promotes membrane protrusion at the leading edge and that RhoA regulates contractility in the cell body. However, there is evidence that RhoA also regulates membrane protrusion. Here we use a fluorescent biosensor, based on a novel design preserving reversible membrane interactions, to visualize the spatiotemporal dynamics of RhoA activity during cell migration. In randomly migrating cells, RhoA activity is concentrated in a sharp band directly at the edge of protrusions. It is observed sporadically in retracting tails, and is low in the cell body. RhoA activity is also associated with peripheral ruffles and pinocytic vesicles, but not with dorsal ruffles induced by platelet-derived growth factor (PDGF). In contrast to randomly migrating cells, PDGF-induced membrane protrusions have low RhoA activity, potentially because PDGF strongly activates Rac, which has previously been shown to antagonize RhoA activity. Our data therefore show that different extracellular cues induce distinct patterns of RhoA signalling during membrane protrusion.


Asunto(s)
Movimiento Celular , Extensiones de la Superficie Celular/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Células 3T3 , Animales , Técnicas Biosensibles , Extensiones de la Superficie Celular/efectos de los fármacos , Fibroblastos , Ratones , Pinocitosis , Factor de Crecimiento Derivado de Plaquetas/farmacología , Factores de Tiempo , Proteínas de Unión al GTP rac/metabolismo
20.
Eur J Cell Biol ; 101(1): 151197, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34958986

RESUMEN

Metastasis remains the main challenge to overcome for treating ovarian cancers. In this study, we investigate the potential role of the Cdc42 GAP StarD13 in the modulation of cell motility, invasion in ovarian cancer cells. StarD13 depletion does not affect the 2D motility of ovarian cancer cells. More importantly, StarD13 inhibits matrix degradation, invadopodia formation and cell invasion through the inhibition of Cdc42. StarD13 does not localize to mature TKS4-labeled invadopodia that possess matrix degradation ability, while a Cdc42 FRET biosensor, detects Cdc42 activation in these invadopodia. In fact, StarD13 localization and Cdc42 activation appear mutually exclusive in invadopodial structures. Finally, for the first time we uncover a potential role of Cdc42 in the direct recruitment of TKS4 to invadopodia. This study emphasizes the specific role of StarD13 as a narrow spatial regulator of Cdc42, inhibiting invasion, suggesting the suitability of StarD13 for targeted therapy.


Asunto(s)
Adenocarcinoma , Proteínas Activadoras de GTPasa/genética , Podosomas , Proteínas Supresoras de Tumor/genética , Proteína de Unión al GTP cdc42/genética , Línea Celular Tumoral , Humanos , Invasividad Neoplásica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA