Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Biochem ; 175(1): 57-67, 2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-37812440

RESUMEN

The Bin-Amphiphysin-Rvs (BAR) domain of endophilin binds to the cell membrane and shapes it into a tubular shape for endocytosis. Endophilin has a Src-homology 3 (SH3) domain at their C-terminal. The SH3 domain interacts with the proline-rich motif (PRM) that is found in proteins such as neural Wiskott-Aldrich syndrome protein (N-WASP). Here, we re-examined the binding sites of the SH3 domain of endophilin in N-WASP by machine learning-based prediction and identified the previously unrecognized binding site. In addition to the well-recognized PRM at the central proline-rich region, we found a PRM in front of the N-terminal WASP homology 1 (WH1) domain of N-WASP (NtPRM) as a binding site of the endophilin SH3 domain. Furthermore, the diameter of the membrane tubules in the presence of NtPRM mutant was narrower and wider than that in the presence of N-WASP and in its absence, respectively. Importantly, the NtPRM of N-WASP was involved in the membrane localization of endophilin A2 in cells. Therefore, the NtPRM contributes to the binding of endophilin to N-WASP in membrane remodeling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras , Proteínas Portadoras/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Sitios de Unión , Dominios Homologos src , Factores de Transcripción/metabolismo , Prolina/metabolismo , Unión Proteica
2.
Sci Adv ; 9(17): eadf5143, 2023 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-37126564

RESUMEN

The higher-order assembly of Bin-amphiphysin-Rvs (BAR) domain proteins, including the FCH-BAR (F-BAR) domain proteins, into lattice on the membrane is essential for the formation of subcellular structures. However, the regulation of their ordered assembly has not been elucidated. Here, we show that the higher ordered assembly of growth-arrested specific 7 (GAS7), an F-BAR domain protein, is regulated by the multivalent scaffold proteins of Wiskott-Aldrich syndrome protein (WASP)/neural WASP, that commonly binds to the BAR domain superfamily proteins, together with WISH, Nck, the activated small guanosine triphosphatase Cdc42, and a membrane-anchored phagocytic receptor. The assembly kinetics by fluorescence resonance energy transfer monitoring indicated that the GAS7 assembly on liposomes started within seconds and was further increased by the presence of these proteins. The regulated GAS7 assembly was abolished by Wiskott-Aldrich syndrome mutations both in vitro and in cellular phagocytosis. Therefore, Cdc42 and the scaffold proteins that commonly bind to the BAR domain superfamily proteins promoted GAS7 assembly.


Asunto(s)
Proteínas de Unión al GTP Monoméricas , Proteína del Síndrome de Wiskott-Aldrich , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteína Neuronal del Síndrome de Wiskott-Aldrich/genética , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Actinas/metabolismo
3.
Adv Sci (Weinh) ; 10(7): e2207368, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36698307

RESUMEN

The cell migration cycle, well-established in 2D, proceeds with forming new protrusive structures at the cell membrane and subsequent redistribution of contractile machinery. Three-dimensional (3D) environments are complex and composed of 1D fibers, and 1D fibers are shown to recapitulate essential features of 3D migration. However, the establishment of protrusive activity at the cell membrane and contractility in 1D fibrous environments remains partially understood. Here the role of membrane curvature regulator IRSp53 is examined as a coupler between actin filaments and plasma membrane during cell migration on single, suspended 1D fibers. IRSp53 depletion reduced cell-length spanning actin stress fibers that originate from the cell periphery, protrusive activity, and contractility, leading to uncoupling of the nucleus from cellular movements. A theoretical model capable of predicting the observed transition of IRSp53-depleted cells from rapid stick-slip migration to smooth and slower migration due to reduced actin polymerization at the cell edges is developed, which is verified by direct measurements of retrograde actin flow using speckle microscopy. Overall, it is found that IRSp53 mediates actin recruitment at the cellular tips leading to the establishment of cell-length spanning fibers, thus demonstrating a unique role of IRSp53 in controlling cell migration in 3D.


Asunto(s)
Citoesqueleto de Actina , Actinas , Movimiento Celular , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Membrana Celular/metabolismo , Movimiento Celular/genética , Núcleo Celular/metabolismo , Seudópodos/genética , Seudópodos/metabolismo
5.
PLoS One ; 17(7): e0271003, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35834519

RESUMEN

Caveolae are plasma membrane invaginations that play important roles in both endocytosis and membrane tension buffering. Typical caveolae have invaginated structures with a high-density caveolin assembly. Membrane sculpting proteins, including PACSIN2 and EHD2, are involved in caveolar biogenesis. PACSIN2 is an F-BAR domain-containing protein with a membrane sculpting ability that is essential for caveolar shaping. EHD2 is also localized at caveolae and involved in their stability. However, the spatial relationship between PACSIN2, EHD2, and caveolin has not yet been investigated. We observed the single-molecule localizations of PACSIN2 and EHD2 relative to caveolin-1 in three-dimensional space. The single-molecule localizations were grouped by their proximity localizations into the geometric structures of blobs. In caveolin-1 blobs, PACSIN2, EHD2, and caveolin-1 had overlapped spatial localizations. Interestingly, the mean centroid of the PACSIN2 F-BAR domain at the caveolin-1 blobs was closer to the plasma membrane than those of EHD2 and caveolin-1, suggesting that PACSIN2 is involved in connecting caveolae to the plasma membrane. Most of the blobs with volumes typical of caveolae had PACSIN2 and EHD2, in contrast to those with smaller volumes. Therefore, PACSIN2 and EHD2 are apparently localized at typically sized caveolae.


Asunto(s)
Caveolas , Caveolina 1 , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Caveolas/metabolismo , Caveolina 1/metabolismo , Membrana Celular/metabolismo , Endocitosis , Proteínas de la Membrana/metabolismo
6.
Mol Biol Cell ; 32(21): br7, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34495704

RESUMEN

Superresolution microscopy determines the localization of fluorescent proteins with high precision, beyond the diffraction limit of light. Superresolution microscopic techniques include photoactivated localization microscopy (PALM), which can localize a single protein by the stochastic activation of its fluorescence. In the determination of single-molecule localization by PALM, the number of molecules that can be analyzed per image is limited. Thus, many images are required to reconstruct the localization of numerous molecules in the cell. However, most fluorescent proteins lose their fluorescence upon fixation. Here, we combined the amino acid substitutions of two Eos protein derivatives, Skylan-S and mEos4b, which are a green reversibly photoswitchable fluorescent protein (RSFP) and a fixation-resistant green-to-red photoconvertible fluorescent protein, respectively, resulting in the fixation-resistant Skylan-S (frSkylan-S), a green RSFP. The frSkylan-S protein is inactivated by excitation light and reactivated by irradiation with violet light, and retained more fluorescence after aldehyde fixation than Skylan-S. The qualities of the frSkylan-S fusion proteins were sufficiently high in PALM observations, as examined using α-tubulin and clathrin light chain. Furthermore, frSkylan-S can be combined with antibody staining for multicolor imaging. Therefore, frSkylan-S is a green fluorescent protein suitable for PALM imaging under aldehyde-fixation conditions.


Asunto(s)
Microscopía Fluorescente/métodos , Imagen Individual de Molécula/métodos , Animales , Proteínas de Unión al ADN/genética , Colorantes Fluorescentes/química , Proteínas Fluorescentes Verdes/química , Proteínas Luminiscentes/química , Ratones , Proteínas del Tejido Nervioso/genética , Proteína Fluorescente Roja
7.
Front Cell Dev Biol ; 9: 635231, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34422790

RESUMEN

Protein localization in cells has been analyzed by fluorescent labeling using indirect immunofluorescence and fluorescent protein tagging. However, the relationships between the localization of different proteins had not been analyzed using artificial intelligence. Here, we applied convolutional networks for the prediction of localization of the cytoskeletal proteins from the localization of the other proteins. Lamellipodia are one of the actin-dependent subcellular structures involved in cell migration and are mainly generated by the Wiskott-Aldrich syndrome protein (WASP)-family verprolin homologous protein 2 (WAVE2) and the membrane remodeling I-BAR domain protein IRSp53. Focal adhesion is another actin-based structure that contains vinculin protein and promotes lamellipodia formation and cell migration. In contrast, microtubules are not directly related to actin filaments. The convolutional network was trained using images of actin filaments paired with WAVE2, IRSp53, vinculin, and microtubules. The generated images of WAVE2, IRSp53, and vinculin were highly similar to their real images. In contrast, the microtubule images generated from actin filament images were inferior without the generation of filamentous structures, suggesting that microscopic images of actin filaments provide more information about actin-related protein localization. Collectively, this study suggests that image translation by the convolutional network can predict the localization of functionally related proteins, and the convolutional network might be used to describe the relationships between the proteins by their localization.

8.
STAR Protoc ; 2(3): 100625, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34223199

RESUMEN

Extracellular vesicles (EVs) play important roles in extracellular trafficking and signaling. Here, we separate EVs by differential centrifugation. EVs separated by this approach are called large EVs (l-EVs) and small EVs (s-EVs), reflecting particle size, which sediment based on different ultracentrifugation forces. The resulting EVs can be quantified and analyzed using nanoparticle tracking analysis, immunoblotting, and functional assays. This protocol was applied to a suspension cell line with high transfection efficiency adapted to a high-density, serum-free culture. For complete details on the use and execution of this protocol, please refer to Nishimura et al. (2021).


Asunto(s)
Vesículas Extracelulares/metabolismo , Ultracentrifugación/métodos , Western Blotting , Movimiento Celular , Medio de Cultivo Libre de Suero , Células HEK293 , Humanos , Proteínas/aislamiento & purificación
9.
Dev Cell ; 56(6): 842-859.e8, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33756122

RESUMEN

Extracellular vesicles (EVs) are classified as large EVs (l-EVs, or microvesicles) and small EVs (s-EVs, or exosomes). S-EVs are thought to be generated from endosomes through a process that mainly depends on the ESCRT protein complex, including ALG-2 interacting protein X (ALIX). However, the mechanisms of l-EV generation from the plasma membrane have not been identified. Membrane curvatures are generated by the bin-amphiphysin-rvs (BAR) family proteins, among which the inverse BAR (I-BAR) proteins are involved in filopodial protrusions. Here, we show that the I-BAR proteins, including missing in metastasis (MIM), generate l-EVs by scission of filopodia. Interestingly, MIM-containing l-EV production was promoted by in vivo equivalent external forces and by the suppression of ALIX, suggesting an alternative mechanism of vesicle formation to s-EVs. The MIM-dependent l-EVs contained lysophospholipids and proteins, including IRS4 and Rac1, which stimulated the migration of recipient cells through lamellipodia formation. Thus, these filopodia-dependent l-EVs, which we named as filopodia-derived vesicles (FDVs), modify cellular behavior.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Movimiento Celular , Endocitosis , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Exosomas/metabolismo , Vesículas Extracelulares/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas de Neoplasias/metabolismo , Seudópodos/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/genética , Membrana Celular/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Células HEK293 , Humanos , Proteínas de Microfilamentos/genética , Proteínas de Neoplasias/genética
10.
Nat Commun ; 10(1): 4763, 2019 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-31628328

RESUMEN

Phagocytosis is a cellular process for internalization of micron-sized large particles including pathogens. The Bin-Amphiphysin-Rvs167 (BAR) domain proteins, including the FCH-BAR (F-BAR) domain proteins, impose specific morphologies on lipid membranes. Most BAR domain proteins are thought to form membrane invaginations or protrusions by assembling into helical submicron-diameter filaments, such as on clathrin-coated pits, caveolae, and filopodia. However, the mechanism by which BAR domain proteins assemble into micron-scale phagocytic cups was unclear. Here, we show that the two-dimensional sheet-like assembly of Growth Arrest-Specific 7 (GAS7) plays a critical role in phagocytic cup formation in macrophages. GAS7 has the F-BAR domain that possesses unique hydrophilic loops for two-dimensional sheet formation on flat membranes. Super-resolution microscopy reveals the similar assemblies of GAS7 on phagocytic cups and liposomes. The mutations of the loops abolishes both the membrane localization of GAS7 and phagocytosis. Thus, the sheet-like assembly of GAS7 plays a significant role in phagocytosis.


Asunto(s)
Macrófagos/metabolismo , Lípidos de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fagocitosis , Secuencia de Aminoácidos , Animales , Membrana Celular/química , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Células HeLa , Humanos , Lípidos de la Membrana/química , Ratones , Microscopía Electrónica , Microscopía Fluorescente , Modelos Moleculares , Mutación , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Dominios Proteicos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Células RAW 264.7 , Homología de Secuencia de Aminoácido
11.
Biochem Soc Trans ; 46(2): 379-389, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29540508

RESUMEN

Lipid membranes are structural components of cell surfaces and intracellular organelles. Alterations in lipid membrane shape are accompanied by numerous cellular functions, including endocytosis, intracellular transport, and cell migration. Proteins containing Bin-Amphiphysin-Rvs (BAR) domains (BAR proteins) are unique, because their structures correspond to the membrane curvature, that is, the shape of the lipid membrane. BAR proteins present at high concentration determine the shape of the membrane, because BAR domain oligomers function as scaffolds that mould the membrane. BAR proteins co-operate with various molecular and non-molecular factors. The molecular factors include cytoskeletal proteins such as the regulators of actin filaments and the membrane scission protein dynamin. Lipid composition, including saturated or unsaturated fatty acid tails of phospholipids, also affects the ability of BAR proteins to mould the membrane. Non-molecular factors include the external physical forces applied to the membrane, such as tension and friction. In this mini-review, we will discuss how the BAR proteins orchestrate membrane dynamics together with various molecular and non-molecular factors.


Asunto(s)
Lípidos de la Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/metabolismo , Lípidos de la Membrana/química , Proteínas de la Membrana/química , Simulación de Dinámica Molecular , Proteínas del Tejido Nervioso/química , Polimerizacion , Dominios Proteicos
12.
Nat Commun ; 8(1): 1834, 2017 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-29184140

RESUMEN

Normal epithelial cells are stably connected to each other via the apical junctional complex (AJC). AJCs, however, tend to be disrupted during tumor progression, and this process is implicated in cancer dissemination. Here, using colon carcinoma cells that fail to form AJCs, we investigated molecular defects behind this failure through a search for chemical compounds that could restore AJCs, and found that microtubule-polymerization inhibitors (MTIs) were effective. MTIs activated GEF-H1/RhoA signaling, causing actomyosin contraction at the apical cortex. This contraction transmitted force to the cadherin-catenin complex, resulting in a mechanosensitive recruitment of vinculin to cell junctions. This process, in turn, recruited PDZ-RhoGEF to the junctions, leading to the RhoA/ROCK/LIM kinase/cofilin-dependent stabilization of the junctions. RhoGAP depletion mimicked these MTI-mediated processes. Cells that normally organize AJCs did not show such MTI/RhoA sensitivity. Thus, advanced carcinoma cells require elevated RhoA activity for establishing robust junctions, which triggers tension-sensitive reorganization of actin/adhesion regulators.


Asunto(s)
Uniones Adherentes/metabolismo , Moléculas de Adhesión Celular/metabolismo , Adhesión Celular/fisiología , Neoplasias del Colon/metabolismo , Proteínas del Citoesqueleto/metabolismo , Células HT29/fisiología , Uniones Intercelulares/fisiología , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestructura , Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Uniones Adherentes/ultraestructura , Fenómenos Biomecánicos , Células CACO-2 , Cadherinas/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/patología , Células Epiteliales/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HT29/citología , Células HT29/efectos de los fármacos , Humanos , Quinasas Lim/metabolismo , Microtúbulos , Miosina Tipo II/metabolismo , Nocodazol/farmacología , Transducción de Señal , Vinculina/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
13.
J Cell Biol ; 215(4): 559-573, 2016 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-27807130

RESUMEN

Epithelial junctions comprise two subdomains, the apical junctional complex (AJC) and the adjacent lateral membrane contacts (LCs), that span the majority of the junction. The AJC is lined with circumferential actin cables, whereas the LCs are associated with less-organized actin filaments whose roles are elusive. We found that DAAM1, a formin family actin regulator, accumulated at the LCs, and its depletion caused dispersion of actin filaments at these sites while hardly affecting circumferential actin cables. DAAM1 loss enhanced the motility of LC-forming membranes, leading to their invasion of neighboring cell layers, as well as disruption of polarized epithelial layers. We found that components of the WAVE complex and its downstream targets were required for the elevation of LC motility caused by DAAM1 loss. These findings suggest that the LC membranes are motile by nature because of the WAVE complex, but DAAM1-mediated actin regulation normally restrains this motility, thereby stabilizing epithelial architecture, and that DAAM1 loss evokes invasive abilities of epithelial cells.


Asunto(s)
Membrana Celular/metabolismo , Células Epiteliales/metabolismo , Uniones Intercelulares/metabolismo , Proteínas de Microfilamentos/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Actinas/metabolismo , Animales , Cadherinas/metabolismo , Extensiones de la Superficie Celular/metabolismo , Células HEK293 , Humanos , Ratones , Transducción de Señal , alfa Catenina/metabolismo , beta Catenina/metabolismo , Proteínas de Unión al GTP rac/metabolismo
14.
Cell ; 149(5): 1084-97, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22632972

RESUMEN

Neural-tube closure is a critical step of embryogenesis, and its failure causes serious birth defects. Coordination of two morphogenetic processes--convergent extension and neural-plate apical constriction--ensures the complete closure of the neural tube. We now provide evidence that planar cell polarity (PCP) signaling directly links these two processes. In the bending neural plates, we find that a PCP-regulating cadherin, Celsr1, is concentrated in adherens junctions (AJs) oriented toward the mediolateral axes of the plates. At these AJs, Celsr1 cooperates with Dishevelled, DAAM1, and the PDZ-RhoGEF to upregulate Rho kinase, causing their actomyosin-dependent contraction in a planar-polarized manner. This planar-polarized contraction promotes simultaneous apical constriction and midline convergence of neuroepithelial cells. Together our findings demonstrate that PCP signals confer anisotropic contractility on the AJs, producing cellular forces that promote the polarized bending of the neural plate.


Asunto(s)
Polaridad Celular , Embrión de Pollo/metabolismo , Morfogénesis , Tubo Neural/metabolismo , Uniones Adherentes/metabolismo , Animales , Línea Celular , Perros , Humanos , Ratones , Placa Neural/metabolismo
15.
Biochem Biophys Res Commun ; 398(2): 217-23, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20599736

RESUMEN

Centrosome cycle is strictly coordinated with chromosome duplication cycle to ensure the faithful segregation of chromosomes. Centrosome duplication occurs from the beginning of S phase, and the duplicated centrosomes are held together by centrosome cohesion to function as a single microtubule organizing center during interphase. At late G2 phase centrosome cohesion is disassembled by Nek2A kinase-mediated phosphorylation and, as a consequence, centrosomes are split and constitute spindle poles in mitosis. It has been reported that depletion of a centrosomal protein kendrin (also named pericentrin) induces premature centrosome splitting in interphase, however, it remains unknown how kendrin contributes to the maintenance of centrosome cohesion. Here we show that kendrin associates with Nek2A kinase, which exhibits considerably low activity. Nek2A kinase activity is inhibited in vitro by addition of the Nek2A-binding region of kendrin in a dose-dependent manner. Furthermore, ectopic expression of the same region decreases the number of the cells with split centrosomes at late G2 phase. Taken together, these results suggest that kendrin anchors Nek2A and suppresses its kinase activity at the centrosomes, and thus, is involved in the mechanism to prevent premature centrosome splitting during interphase.


Asunto(s)
Proteínas de Unión a Calmodulina/metabolismo , Centrosoma/metabolismo , Interfase , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Proteínas de Unión a Calmodulina/genética , Línea Celular , Humanos , Quinasas Relacionadas con NIMA , Proteínas Serina-Treonina Quinasas/genética
16.
Curr Top Dev Biol ; 89: 33-54, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19737641

RESUMEN

Morphogenesis of epithelial tissues involves various forms of reshaping of cell layers, such as invagination or bending, convergent extension, and epithelial-mesenchymal transition. At the cellular level, these processes include changes in the shape, position, and assembly pattern of cells. During such morphogenetic processes, epithelial sheets in general maintain their multicellular architecture, implying that they must engage the mechanisms to change the spatial relationship with their neighbors without disrupting the junctions. A major junctional structure in epithelial tissues is the "adherens junction," which is composed of cadherin adhesion receptors and associated proteins including F-actin. The adherens junctions are required for the firm associations between cells, as disruption of them causes disorganization of the epithelial architecture. The adherens junctions, however, appear to be a dynamic entity, allowing the rearrangement of cells within cell sheets. This dynamic nature of the adherens junctions seems to be supported by various mechanisms, such as the interactions of cadherins with actin cytoskeleton, endocytosis and recycling of cadherins, and the cooperation of cadherins with other adhesion receptors. In this chapter, we provide an overview of these mechanisms analyzed in vitro and in vivo.


Asunto(s)
Uniones Adherentes/metabolismo , Morfogénesis , Uniones Adherentes/enzimología , Animales , Cadherinas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Endocitosis , Humanos , Proteínas de Unión al GTP Monoméricas/metabolismo , Nectinas
17.
Development ; 135(8): 1493-502, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18339671

RESUMEN

Remodeling of epithelial sheets plays important roles in animal morphogenesis. Shroom3 is known to regulate the apical constriction of epithelial cells. Here, we show that Shroom3 binds ROCKs and recruits them to the epithelial apical junctions. We identified the Shroom3-binding site (RII-C1) on ROCKs, and found that RII-C1 could antagonize the Shroom3-ROCK interaction, interfering with the action of Shroom3 on cell morphology. In the invaginating neural plate/tube, Shroom3 colocalized with ROCKs at the apical junctions; Shroom3 depletion or RII-C1 expression in the tube removed these apically localized ROCKs, and concomitantly blocked neural tube closure. Closing neural plate exhibited peculiar cell assemblies, including rosette formation, as well as a planar-polarized distribution of phosphorylated myosin regulatory light chain, but these were abolished by ROCK inhibition or RII-C1 expression. These results demonstrate that the Shroom3-ROCK interaction is crucial for the regulation of epithelial and neuroepithelial cell arrangement and remodeling.


Asunto(s)
Uniones Intercelulares/metabolismo , Proteínas de Microfilamentos/metabolismo , Quinasas Asociadas a rho/metabolismo , Animales , Sitios de Unión , Células COS , Línea Celular , Embrión de Pollo , Chlorocebus aethiops , Perros , Células Epiteliales/metabolismo , Femenino , Ratones , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/genética , Tubo Neural/citología , Tubo Neural/embriología , Tubo Neural/metabolismo , Células Neuroepiteliales/metabolismo , Embarazo , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Quinasas Asociadas a rho/química
18.
Genes Cells ; 10(1): 75-86, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15670215

RESUMEN

Centrosome duplication occurs once per cell cycle and is thought to be triggered by cyclin E-cdk2. However, it is largely unknown how the duplication is regulated. Here, we found that the expression of the centrosome-targeting region of CG-NAP (centrosome and Golgi-localized PKN-associated protein), which we designate as CG-NAP/D, increased the number of centrosomes in Chinese hamster ovary (CHO)-K1 cells. The amplified centrosomes co-localized with centrosome markers gamma-tubulin, centrin-2 and kendrin as well as endogenous CG-NAP. When CG-NAP/D was dislocated from centrosomes by deleting the centrosome-targeting domain or by fusing with a membrane-targeting sequence, centrosome amplification was suppressed. CG-NAP/D interacted with exogenously expressed cyclin E, which co-localized at centrosomes. The immunoprecipitates of CG-NAP/D exhibited histone H1 kinase activity, suggesting the co-immunoprecipitation of active cyclin-cdk complexes. Furthermore, centrosome fractions prepared from cells expressing CG-NAP/D contained increased amount of cdk2 compared with those from control cells. Centrosome amplification by CG-NAP/D was suppressed by co-expression of a mutant cyclin E unable to interact with cdk2. These results suggest that CG-NAP/D causes centrosome amplification by anchoring excess amount of cyclin E-cdk2 to centrosomes and, possibly, CG-NAP participates in centrosome duplication by recruiting cyclin E-cdk2 to centrosomes in normal cell cycle.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Quinasas CDC2-CDC28/metabolismo , Centrosoma/metabolismo , Ciclina E/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas de Anclaje a la Quinasa A , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Quinasas CDC2-CDC28/genética , Proteínas de Unión a Calmodulina/genética , Proteínas de Unión a Calmodulina/metabolismo , Ciclo Celular/fisiología , Línea Celular , Cricetinae , Ciclina E/genética , Quinasa 2 Dependiente de la Ciclina , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Humanos , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
19.
Biochem Biophys Res Commun ; 314(3): 688-94, 2004 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-14741690

RESUMEN

PKN1 is a fatty acid and Rho-activated serine/threonine protein kinase whose catalytic domain is highly homologous to protein kinase C (PKC) family. In yeast two-hybrid screening for PKN1 binding proteins, we identified tumor necrosis factor alpha (TNFalpha) receptor-associated factor 2 (TRAF2). TRAF2 is one of the major mediators of TNF receptor superfamily transducing TNF signal to various functional targets, including activation of NF-kappaB, JNK, and apoptosis. FLAG-tagged PKN1 was co-immunoprecipitated with endogenous TRAF2 from HEK293 cell lysate, and in vitro binding assay using the deletion mutants of TRAF2 showed that PKN1 directly binds to the TRAF domain of TRAF2. PKN1 has the TRAF2-binding consensus sequences PXQX (S/T) at amino acid residues 580-584 (PIQES), and P580AQ582A mutant was not co-immunoprecipitated with TRAF2. Furthermore, the reduced expression of PKN1 by RNA interference (RNAi) down-regulated TRAF2-induced NF-kappaB activation in HEK293T cells. These results suggest that PKN1 is involved in TRAF2-NF-kappaB signaling pathway.


Asunto(s)
FN-kappa B/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas/fisiología , Secuencia de Aminoácidos , Línea Celular , Regulación hacia Abajo , Genes Reporteros/genética , Vectores Genéticos/genética , Humanos , Luciferasas/metabolismo , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Pruebas de Precipitina , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/genética , Proteínas/metabolismo , Interferencia de ARN , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal/fisiología , Factor 2 Asociado a Receptor de TNF , Transfección , Técnicas del Sistema de Dos Híbridos , Levaduras/genética
20.
J Biochem ; 133(2): 181-7, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12761180

RESUMEN

PKNalpha is a fatty acid- and Rho-activated serine/threonine protein kinase having a catalytic domain homologous to members of the protein kinase C family. Recently it was reported that PKNalpha is involved in the p38 mitogen-activated protein kinase (MAPK) signaling pathway. To date, however, how PKNalpha regulates the p38gamma MAPK signaling pathway is unclear. Here we demonstrate that PKNalpha efficiently phosphorylates MLTKalpha (MLK-like mitogen-activated protein triple kinase), which was recently identified as a MAPK kinase kinase (MAPKKK) for the p38 MAPK cascade. Phosphorylation of MLTKalpha by PKNalpha enhances its kinase activity in vitro. Expression of the kinase-negative mutant of PKNalpha inhibited the mobility shift of MLTKalpha caused by osmotic shock in SDS-PAGE. Furthermore, PKNalpha associates with each member of the p38gamma MAPK signaling pathway (p38gamma, MKK6, and MLTKalpha). These results suggest that PKNalpha functions as not only an upstream activator of MLTKalpha but also a putative scaffold protein for the p38gamma MAPK signaling pathway.


Asunto(s)
Quinasas Quinasa Quinasa PAM/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Tirosina Quinasas/fisiología , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Línea Celular , Humanos , MAP Quinasa Quinasa 6 , Quinasas Quinasa Quinasa PAM/genética , Sistema de Señalización de MAP Quinasas , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutación , Fosforilación , Proteína Quinasa C , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...