Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Syst Biol ; 12(3): 860, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26969729

RESUMEN

The signaling events that drive familial breast cancer (FBC) risk remain poorly understood. While the majority of genomic studies have focused on genetic risk variants, known risk variants account for at most 30% of FBC cases. Considering that multiple genes may influence FBC risk, we hypothesized that a pathway-based strategy examining different data types from multiple tissues could elucidate the biological basis for FBC. In this study, we performed integrated analyses of gene expression and exome-sequencing data from peripheral blood mononuclear cells and showed that cell adhesion pathways are significantly and consistently dysregulated in women who develop FBC. The dysregulation of cell adhesion pathways in high-risk women was also identified by pathway-based profiling applied to normal breast tissue data from two independent cohorts. The results of our genomic analyses were validated in normal primary mammary epithelial cells from high-risk and control women, using cell-based functional assays, drug-response assays, fluorescence microscopy, and Western blotting assays. Both genomic and cell-based experiments indicate that cell-cell and cell-extracellular matrix adhesion processes seem to be disrupted in non-malignant cells of women at high risk for FBC and suggest a potential role for these processes in FBC development.


Asunto(s)
Neoplasias de la Mama/metabolismo , Predisposición Genética a la Enfermedad , Transducción de Señal , Anciano , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Adhesión Celular , Estudios de Cohortes , Femenino , Perfilación de la Expresión Génica , Variación Genética , Humanos , Leucocitos Mononucleares/metabolismo , Persona de Mediana Edad
2.
Proc Natl Acad Sci U S A ; 111(49): 17528-33, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25422436

RESUMEN

Cytoskeletal actin assemblies transmit mechanical stresses that molecular sensors transduce into biochemical signals to trigger cytoskeletal remodeling and other downstream events. How mechanical and biochemical signaling cooperate to orchestrate complex remodeling tasks has not been elucidated. Here, we studied remodeling of contractile actomyosin stress fibers. When fibers spontaneously fractured, they recoiled and disassembled actin synchronously. The disassembly rate was accelerated more than twofold above the resting value, but only when contraction increased the actin density to a threshold value following a time delay. A mathematical model explained this as originating in the increased overlap of actin filaments produced by myosin II-driven contraction. Above a threshold overlap, this mechanical signal is transduced into accelerated disassembly by a mechanism that may sense overlap directly or through associated elastic stresses. This biochemical response lowers the actin density, overlap, and stresses. The model showed that this feedback mechanism, together with rapid stress transmission along the actin bundle, spatiotemporally synchronizes actin disassembly and fiber contraction. Similar actin remodeling kinetics occurred in expanding or contracting intact stress fibers but over much longer timescales. The model accurately described these kinetics, with an almost identical value of the threshold overlap that accelerates disassembly. Finally, we measured resting stress fibers, for which the model predicts constant actin overlap that balances disassembly and assembly. The overlap was indeed regulated, with a value close to that predicted. Our results suggest that coordinated mechanical and biochemical signaling enables extended actomyosin assemblies to adapt dynamically to the mechanical stresses they convey and direct their own remodeling.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Retroalimentación Fisiológica , Actinina/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Animales , Elasticidad , Fibroblastos/citología , Fibroblastos/metabolismo , Ratones , Microscopía , Modelos Teóricos , Contracción Muscular , Transducción de Señal , Factores de Tiempo , Zixina/metabolismo
3.
PLoS Genet ; 9(3): e1003406, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23555310

RESUMEN

A variety of human diseases arise from mutations that alter muscle contraction. Evolutionary conservation allows genetic studies in Drosophila melanogaster to be used to better understand these myopathies and suggest novel therapeutic strategies. Integrin-mediated adhesion is required to support muscle structure and function, and expression of Integrin adhesive complex (IAC) proteins is modulated to adapt to varying levels of mechanical stress within muscle. Mutations in flapwing (flw), a catalytic subunit of myosin phosphatase, result in non-muscle myosin hyperphosphorylation, as well as muscle hypercontraction, defects in size, motility, muscle attachment, and subsequent larval and pupal lethality. We find that moderately elevated expression of the IAC protein PINCH significantly rescues flw phenotypes. Rescue requires PINCH be bound to its partners, Integrin-linked kinase and Ras suppressor 1. Rescue is not achieved through dephosphorylation of non-muscle myosin, suggesting a mechanism in which elevated PINCH expression strengthens integrin adhesion. In support of this, elevated expression of PINCH rescues an independent muscle hypercontraction mutant in muscle myosin heavy chain, Mhc(Samba1). By testing a panel of IAC proteins, we show specificity for PINCH expression in the rescue of hypercontraction mutants. These data are consistent with a model in which PINCH is present in limiting quantities within IACs, with increasing PINCH expression reinforcing existing adhesions or allowing for the de novo assembly of new adhesion complexes. Moreover, in myopathies that exhibit hypercontraction, strategic PINCH expression may have therapeutic potential in preserving muscle structure and function.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Contracción Muscular , Enfermedades Musculares , Factores de Transcripción , Animales , Adhesión Celular/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Regulación de la Expresión Génica , Humanos , Integrinas/genética , Integrinas/metabolismo , Contracción Muscular/genética , Contracción Muscular/fisiología , Enfermedades Musculares/genética , Enfermedades Musculares/fisiopatología , Mutación , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
J Cell Sci ; 125(Pt 13): 3185-94, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22467865

RESUMEN

PINCH, integrin-linked kinase (ILK) and Ras suppressor-1 (RSU-1) are molecular scaffolding proteins that form a physical complex downstream of integrins, and have overlapping roles in cellular adhesion. In Drosophila, PINCH and ILK colocalize in cells and have indistinguishable functions in maintaining wing adhesion and integrin to actin linkage in the muscle. We sought to determine whether the direct physical interaction between PINCH and ILK was essential for their functions using transgenic flies expressing a version of PINCH with a point mutation that disrupts ILK binding (PINCH(Q38A)). We demonstrate that the PINCH-ILK interaction is not required for viability, for integrin-mediated adhesion of the wing or muscle, or for maintaining appropriate localization or levels of either PINCH or ILK. These results suggest alternative modes for PINCH localization, stabilization and linkage to the actin cytoskeleton that are independent of a direct interaction with ILK. Furthermore, we identified a synthetic lethality in flies carrying both the PINCH(Q38A) mutation and a null mutation in the gene encoding RSU-1. This lethality does not result from PINCH mislocalization or destabilization, and illustrates a novel compensatory role for RSU-1 in maintaining viability in flies with compromised PINCH-ILK binding. Taken together, this work highlights the existence of redundant mechanisms in adhesion complex assembly that support integrin function in vivo.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/embriología , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/metabolismo , Animales , Animales Modificados Genéticamente/embriología , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/metabolismo , Western Blotting , Cruzamientos Genéticos , Drosophila/enzimología , Drosophila/genética , Proteínas de Drosophila/genética , Embrión no Mamífero/metabolismo , Embrión no Mamífero/patología , Femenino , Integrinas/genética , Integrinas/metabolismo , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Músculos/citología , Músculos/metabolismo , Mutación Puntual , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Estabilidad Proteica , Factores de Transcripción/genética , Alas de Animales/citología , Alas de Animales/metabolismo
5.
Curr Opin Cell Biol ; 18(5): 524-32, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16908128

RESUMEN

Cell substratum adhesion influences a variety of processes including motility, proliferation and survival. In recent years, it has become clear that there are proteins that are capable of shuttling between cell adhesion zones and the nucleus, providing a mechanism for transcellular coordination and communication. Recent findings have given insight into the physiological signals that trigger trafficking of focal adhesion constituents to the nucleus, where they make diverse contributions to the control of gene expression.


Asunto(s)
Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Uniones Célula-Matriz/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Regulación de la Expresión Génica , Humanos , Integrinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Metaloproteínas/metabolismo , Proteínas Musculares/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Paxillin/genética , Paxillin/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Zixina
6.
Biophys J ; 103(10): 2082-92, 2012 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-23200042

RESUMEN

Actin stress fibers (SFs) are load-bearing and mechanosensitive structures. To our knowledge, the mechanisms that enable SFs to sense and respond to strain have not been fully defined. Acute local strain events can involve a twofold extension of a single SF sarcomere, but how these dramatic local events affect the overall SF architecture is not believed to be understood. Here we have investigated how SF architecture adjusts to episodes of local strain that occur in the cell center. Using fluorescently tagged zyxin to track the borders of sarcomeres, we characterize the dynamics of resting sarcomeres and strain-site sarcomeres. We find that sarcomeres flanking a strain site undergo rapid shortening that directly compensates for the strain-site extension, illustrating lateral communication of mechanical information along the length of a stress fiber. When a strain-site sarcomere extends asymmetrically, its adjacent sarcomeres exhibit a parallel asymmetric shortening response, illustrating that flanking sarcomeres respond to strain magnitude. After extension, strain-site sarcomeres become locations of new sarcomere addition, highlighting mechanical strain as a trigger of sarcomere addition and revealing a, to our knowledge, novel type of SF remodeling. Our findings provide evidence to suggest SF sarcomeres act as strain sensors and are interconnected to support communication of mechanical information.


Asunto(s)
Sarcómeros/metabolismo , Fibras de Estrés/metabolismo , Actinas/metabolismo , Animales , Fenómenos Biomecánicos , Supervivencia Celular , Fibroblastos/citología , Fibroblastos/metabolismo , Homeostasis , Ratones , Modelos Biológicos , Estrés Mecánico
7.
Mol Biol Cell ; 33(11): ar100, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35767320

RESUMEN

The small heat shock protein HspB1, also known as Hsp25/27, is a ubiquitously expressed molecular chaperone that responds to mechanical cues. Uniaxial cyclic stretch activates the p38 mitogen-activated protein kinase (MAPK) signaling cascade and increases the phosphorylation of HspB1. Similar to the mechanosensitive cytoskeletal regulator zyxin, phospho-HspB1 is recruited to features of the stretch-stimulated actin cytoskeleton. To evaluate the role of HspB1 and its phosphoregulation in modulating cell function, we utilized CRISPR/Cas9-edited HspB1-null cells and determined they were altered in behaviors such as actin cytoskeletal remodeling, cell spreading, and cell motility. In our model system, expression of WT HspB1, but not nonphosphorylatable HspB1, rescued certain characteristics of the HspB1-null cells including the enhanced cell motility of HspB1-null cells and the deficient actin reinforcement of stretch-stimulated HspB1-null cells. The recruitment of HspB1 to high-tension structures in geometrically constrained cells, such as actin comet tails emanating from focal adhesions, also required a phosphorylatable HspB1. We show that mechanical signals activate posttranslational regulation of the molecular chaperone, HspB1, and are required for normal cell behaviors including actin cytoskeletal remodeling, cell spreading, and cell migration.


Asunto(s)
Actinas , Proteínas de Choque Térmico Pequeñas , Actinas/metabolismo , Movimiento Celular/fisiología , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas de Choque Térmico Pequeñas/metabolismo , Chaperonas Moleculares/metabolismo , Fosforilación
8.
Heliyon ; 8(12): e12147, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36619427

RESUMEN

Formation of robust actomyosin stress fibers (SF) in response to cell stretch plays a key role in the transfer of information from the cytoplasm into the nucleus. Actin/LINC/Lamin (ALL) nuclear lines provide mechanical linkage between the actin cytoskeleton and the lamin nucleoskeleton across the nuclear envelope. To understand the establishment of ALL lines, we used live cell imaging of cells exposed to cyclic stretch. We discovered that nuclear pore complexes (NPCs) concentrate along ALL lines that are generated in response to uniaxial cyclic stretch. The ALL-associated NPCs display increased fluorescence intensity of nucleoporins Pom121, TPR and Nup153 relative to nucleoporins that are distal to the ALL lines. Here we test the hypothesis that a LINC complex component of ALL lines, SUN1 is involved in the integration of NPCs with ALL lines. We generated CRISPR SUN1 knockdown and knockout cell lines and show that SUN1 is essential for normal integration of NPCs to ALL lines. Loss or elimination of SUN1 significantly diminishes NPC/ALL line integration, demonstrating a key role for SUN1 in the recruitment or stabilization of NPCs to a discrete subdomain of the nuclear envelope at ALL lines. This work provides new insight into the mechanism by which cells respond to mechanical force through nuclear envelope remodeling.

9.
Am J Physiol Cell Physiol ; 301(2): C373-82, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21562304

RESUMEN

Muscle LIM protein (MLP) can be found at the Z-disk of sarcomeres where it is hypothesized to be involved in sensing muscle stretch. Loss of murine MLP results in dilated cardiomyopathy, and mutations in human MLP lead to cardiac hypertrophy, indicating a critical role for MLP in maintaining normal cardiac function. Loss of MLP in Drosophila (mlp84B) also leads to muscle dysfunction, providing a model system to examine MLP's mechanism of action. Mlp84B-null flies that survive to adulthood are not able to fly or beat their wings. Transgenic expression of the mlp84B gene in the Mlp84B-null background rescues flight ability and restores wing beating ability. Mechanical analysis of skinned flight muscle fibers showed a 30% decrease in oscillatory power production and a slight increase in the frequency at which maximum power is generated for fibers lacking Mlp84B compared with rescued fibers. Mlp84B-null muscle fibers displayed a 25% decrease in passive, active, and rigor stiffness compared with rescued fibers, but no significant decrease in isometric tension generation was observed. Muscle ultrastructure of Mlp84B-null muscle fibers is grossly normal; however, the null fibers have a slight decrease, 11%, in thick filament number per unit cross-sectional area. Our data indicate that MLP contributes to muscle stiffness and is necessary for maximum work and power generation.


Asunto(s)
Proteínas de Drosophila/deficiencia , Drosophila/metabolismo , Vuelo Animal , Eliminación de Gen , Contracción Isométrica , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/deficiencia , Fuerza Muscular , Alas de Animales/metabolismo , Análisis de Varianza , Animales , Fenómenos Biomecánicos , Drosophila/genética , Drosophila/ultraestructura , Proteínas de Drosophila/genética , Genotipo , Proteínas con Dominio LIM , Microscopía Electrónica de Transmisión , Fibras Musculares Esqueléticas/ultraestructura , Proteínas Musculares/genética , Fenotipo , Alas de Animales/ultraestructura
10.
Curr Opin Cell Biol ; 14(1): 88-103, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11792550

RESUMEN

Actin filament assembly is a tightly regulated process that functions in many aspects of cell physiology. Members of the Ena/VASP (Drosophila Enabled/vasodilator-stimulated phosphoprotein) family are key players in regulating actin filament assembly, in many cases through their association with binding partners that display a particular proline-rich motif, FPPPP. Ena/VASP proteins interact with these partners via the highly conserved Ena/VASP homology 1 (EVH1) domain. The diverse array of binding partners for EVH1 domains, including cytoskeletal proteins such as zyxin, transmembrane guidance receptors such as Roundabout, and the T-cell signaling protein Fyb/SLAP, shows that these interactions are likely to be important in a number of cellular processes that require regulated actin filament assembly.


Asunto(s)
Movimiento Celular , Polaridad Celular , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/fisiología , Secuencias de Aminoácidos , Animales , Moléculas de Adhesión Celular/química , Moléculas de Adhesión Celular/metabolismo , Moléculas de Adhesión Celular/fisiología , Epitelio/crecimiento & desarrollo , Proteínas de Microfilamentos/metabolismo , Modelos Biológicos , Modelos Moleculares , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiología , Prolina/metabolismo , Estructura Terciaria de Proteína
11.
J Cell Biol ; 172(5): 771-82, 2006 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-16505170

RESUMEN

Focal adhesions are specialized regions of the cell surface where integrin receptors and associated proteins link the extracellular matrix to the actin cytoskeleton. To define the cellular role of the focal adhesion protein zyxin, we characterized the phenotype of fibroblasts in which the zyxin gene was deleted by homologous recombination. Zyxin-null fibroblasts display enhanced integrin-dependent adhesion and are more migratory than wild-type fibroblasts, displaying reduced dependence on extracellular matrix cues. We identified differences in the profiles of 75- and 80-kD tyrosine-phosphorylated proteins in the zyxin-null cells. Tandem array mass spectrometry identified both modified proteins as isoforms of the actomyosin regulator caldesmon, a protein known to influence contractility, stress fiber formation, and motility. Zyxin-null fibroblasts also show deficits in actin stress fiber remodeling and exhibit changes in the molecular composition of focal adhesions, most notably by severely reduced accumulation of Ena/VASP proteins. We postulate that zyxin cooperates with Ena/VASP proteins and caldesmon to influence integrin-dependent cell motility and actin stress fiber remodeling.


Asunto(s)
Actinas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular/genética , Proteínas del Citoesqueleto/metabolismo , Metaloproteínas/deficiencia , Metaloproteínas/genética , Proteínas de Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Actinas/deficiencia , Animales , Proteínas de Unión a Calmodulina/metabolismo , Adhesión Celular/genética , Línea Celular Transformada , Células Cultivadas , Depsipéptidos/farmacología , Matriz Extracelular/fisiología , Fibroblastos/metabolismo , Integrinas/biosíntesis , Integrinas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitosis/fisiología , Fibras de Estrés/efectos de los fármacos , Zixina
12.
J Cell Biol ; 174(3): 447-58, 2006 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-16880273

RESUMEN

Cell-substrate contacts, called focal adhesions (FAs), are dynamic in rapidly moving cells. We show that supervillin (SV)--a peripheral membrane protein that binds myosin II and F-actin in such cells--negatively regulates stress fibers, FAs, and cell-substrate adhesion. The major FA regulatory sequence within SV (SV342-571) binds to the LIM domains of two proteins in the zyxin family, thyroid receptor-interacting protein 6 (TRIP6) and lipoma-preferred partner (LPP), but not to zyxin itself. SV and TRIP6 colocalize within large FAs, where TRIP6 may help recruit SV. RNAi-mediated decreases in either protein increase cell adhesion to fibronectin. TRIP6 partially rescues SV effects on stress fibers and FAs, apparently by mislocating SV away from FAs. Thus, SV interactions with TRIP6 at FAs promote loss of FA structure and function. SV and TRIP6 binding partners suggest several specific mechanisms through which the SV-TRIP6 interaction may regulate FA maturation and/or disassembly.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adhesiones Focales/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Factores de Transcripción/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Proteínas Adaptadoras Transductoras de Señales/química , Animales , Células COS , Bovinos , Células Cultivadas , Chlorocebus aethiops , Regulación hacia Abajo/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Proteínas con Dominio LIM , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Miocitos del Músculo Liso/citología , Proteínas Nucleares/metabolismo , Complejo de la Endopetidasa Proteasomal , Unión Proteica , Ratas , Secuencias Reguladoras de Ácidos Nucleicos/genética , Factores de Transcripción/química , Región del Complejo T del Genoma
13.
Arterioscler Thromb Vasc Biol ; 30(4): 694-701, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20056913

RESUMEN

OBJECTIVE: Cysteine-rich protein (CRP) 1 and 2 are cytoskeletal lin-11 isl-1 mec-3 (LIM)-domain proteins thought to be critical for smooth muscle differentiation. Loss of murine CRP2 does not overtly affect smooth muscle differentiation or vascular function but does exacerbate neointima formation in response to vascular injury. Because CRPs 1 and 2 are coexpressed in the vasculature, we hypothesize that CRPs 1 and 2 act redundantly in smooth muscle differentiation. METHODS AND RESULTS: We generated Csrp1 (gene name for CRP1) null mice by genetic ablation of the Csrp1 gene and found that mice lacking CRP1 are viable and fertile. Smooth muscle-containing tissues from Csrp1-null mice are morphologically indistinguishable from wild-type mice and have normal contractile properties. Mice lacking CRPs 1 and 2 are viable and fertile, ruling out functional redundancy between these 2 highly related proteins as a cause for the lack of an overt phenotype in the Csrp1-null mice. Csrp1-null mice challenged by wire-induced arterial injury display reduced neointima formation, opposite to that seen in Csrp2-null mice, whereas Csrp1/Csrp2 double-null mice produce a wild-type response. CONCLUSIONS: Smooth muscle CRPs are not essential for normal smooth muscle differentiation during development, but may act antagonistically to modulate the smooth muscle response to pathophysiological stress.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Nucleares/deficiencia , Túnica Íntima/metabolismo , Animales , Apoptosis , Movimiento Celular , Células Cultivadas , Arteria Femoral/metabolismo , Arteria Femoral/patología , Genotipo , Hiperplasia , Proteínas con Dominio LIM , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/patología , Proteínas Nucleares/genética , Fenotipo , Factores de Tiempo , Transfección , Túnica Íntima/lesiones , Túnica Íntima/patología , Vasoconstricción
14.
FASEB J ; 23(3): 916-28, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19017743

RESUMEN

We recently established the critical role of the PTEN/MAGI-1b signalosome in stabilization of cell-cell contacts and suppression of invasiveness. The PTEN tumor suppressor is recruited to E-cadherin junctional complexes through the binding to the second PDZ domain of the MAGI-1b scaffolding molecule, whereas beta-catenin interacts with the fifth PDZ domain. To identify additional effectors of this signalosome, we used yeast 2-hybrid screening. Among the clones identified, we focused on TRIP6, which belongs to the zyxin family of proteins. We demonstrated that TRIP6 interacted directly with MAGI-1b by binding to its fifth PDZ domain. Ectopic expression of TRIP6 induced invasiveness in the epithelial MDCK and MDCKts-src cells in a PI3-kinase- and a NF-kappaB-dependent manner and impaired cell-cell aggregation at least in part by uncoupling adherens junctional complexes from the cytoskeleton. The TRIP6Stop473 mutant, which lacks the PDZ binding motif, was still able to increase NF-kappaB and Akt activities but did not promote invasiveness or interfere with cell-cell aggregation. Intracellular delivery of competing peptides corresponding to TRIP6 or beta-catenin C terminus restored invasive properties in MDCKts-src TRIP6Stop473 cells, highlighting the requirement of PDZ scaffolds in junctional complexes activity. TRIP6 overexpression in colon tumors suggest its critical role in cancer progression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Factores de Transcripción/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células CACO-2 , Cadherinas/metabolismo , Adhesión Celular , Moléculas de Adhesión Celular , Moléculas de Adhesión Celular Neuronal/genética , Línea Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Citoesqueleto/metabolismo , Perros , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Guanilato-Quinasas , Células HeLa , Humanos , Proteínas con Dominio LIM , FN-kappa B/metabolismo , Complejo de la Endopetidasa Proteasomal , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción/genética , Transfección , Técnicas del Sistema de Dos Híbridos , Proteínas de Unión al GTP rho/metabolismo
15.
J Cell Biol ; 171(2): 209-15, 2005 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-16247023

RESUMEN

Organs and tissues adapt to acute or chronic mechanical stress by remodeling their actin cytoskeletons. Cells that are stimulated by cyclic stretch or shear stress in vitro undergo bimodal cytoskeletal responses that include rapid reinforcement and gradual reorientation of actin stress fibers; however, the mechanism by which cells respond to mechanical cues has been obscure. We report that the application of either unidirectional cyclic stretch or shear stress to cells results in robust mobilization of zyxin from focal adhesions to actin filaments, whereas many other focal adhesion proteins and zyxin family members remain at focal adhesions. Mechanical stress also induces the rapid zyxin-dependent mobilization of vasodilator-stimulated phosphoprotein from focal adhesions to actin filaments. Thickening of actin stress fibers reflects a cellular adaptation to mechanical stress; this cytoskeletal reinforcement coincides with zyxin mobilization and is abrogated in zyxin-null cells. Our findings identify zyxin as a mechanosensitive protein and provide mechanistic insight into how cells respond to mechanical cues.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Citoesqueleto/metabolismo , Adhesiones Focales/fisiología , Glicoproteínas/metabolismo , Metaloproteínas/metabolismo , Animales , Proteínas del Citoesqueleto , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Humanos , Ratones , Estrés Mecánico , Zixina
16.
Mol Biol Cell ; 18(5): 1723-33, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17332502

RESUMEN

In this report, an antisense RNA strategy has allowed us to show that disruption of ALP expression affects the expression of the muscle transcription factors myogenin and MyoD, resulting in the inhibition of muscle differentiation. Introduction of a MyoD expression construct into ALP-antisense cells is sufficient to restore the capacity of the cells to differentiate, illustrating that ALP function occurs upstream of MyoD. It is known that MyoD is under the control of serum response factor (SRF), a transcriptional regulator whose activity is modulated by actin dynamics. A dramatic reduction of actin filament bundles is observed in ALP-antisense cells and treatment of these cells with the actin-stabilizing drug jasplakinolide stimulates SRF activity and restores the capacity of the cells to differentiate. Furthermore, we show that modulation of ALP expression influences SRF activity, the level of its coactivator, MAL, and muscle differentiation. Collectively, these results suggest a critical role of ALP on muscle differentiation, likely via cytoskeletal regulation of SRF.


Asunto(s)
Proteínas de Microfilamentos/metabolismo , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/metabolismo , Factor de Respuesta Sérica/metabolismo , Actinina/metabolismo , Actinas/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular , Línea Celular , Citoesqueleto/metabolismo , Cartilla de ADN/genética , Expresión Génica , Proteínas con Dominio LIM , Ratones , Proteínas de Microfilamentos/antagonistas & inhibidores , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Desarrollo de Músculos/genética , Desarrollo de Músculos/fisiología , Proteína MioD/genética , Proteína MioD/metabolismo , Factor 5 Regulador Miogénico/genética , Factor 5 Regulador Miogénico/metabolismo , ARN sin Sentido/genética , Transfección
17.
HPB (Oxford) ; 12(5): 352-8, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20590912

RESUMEN

OBJECTIVE: Prognostic markers for pancreatic ductal adenocarcinoma (PDA) have failed to accurately predict patient prognosis. Recently, interest has developed in the accuracy of integrin-associated PINCH protein expression in human cancers as a predictive marker of tumour status. The goal of this study was to define the expression of PINCH protein in PDA. METHODS: Human PDA samples and orthotopic tumours from a murine model were analysed by immunohistochemistry for PINCH expression. In the animal model, PINCH expression was compared between primary and metastatic tumours. In the human samples, PINCH expression was correlated with stage, nodal involvement, margin status and overall survival. RESULTS: In the murine model, there was greater PINCH expression in metastatic tumours than in primary tumours. In the human PDA samples, greater staining for PINCH in the tumour cells was correlated with higher T status. Additionally, high PINCH expression in the stroma was associated with decreased overall survival. CONCLUSIONS: Findings of increased PINCH protein in more advanced stages of human PDA, as well as in metastatic tumours in the animal model, support the hypothesis that PINCH is an important controller of cell survival and migration. Additionally, the importance of the differential expression of PINCH in the human tumour and stroma warrants further evaluation.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Proteínas de Unión al ADN/metabolismo , Neoplasias Pancreáticas/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Anciano , Anciano de 80 o más Años , Animales , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/secundario , Carcinoma Ductal Pancreático/cirugía , Línea Celular Tumoral , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Proteínas con Dominio LIM , Metástasis Linfática , Masculino , Proteínas de la Membrana , Ratones , Ratones Desnudos , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Pancreaticoduodenectomía , Modelos de Riesgos Proporcionales , Medición de Riesgo , Factores de Riesgo , Células del Estroma/metabolismo , Factores de Tiempo , Resultado del Tratamiento , Regulación hacia Arriba
18.
Sci Rep ; 10(1): 19303, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-33168840

RESUMEN

Platelet Derived Growth Factor Receptor (PDGFR) signaling is a central mitogenic pathway in development, as well as tissue repair and homeostasis. The rules governing the binding of PDGF ligand to the receptor to produce activation and downstream signaling have been well defined over the last several decades. In cultured cells after a period of serum deprivation, treatment with PDGF leads to the rapid formation of dramatic, actin-rich Circular Dorsal Ruffles (CDRs). Using CDRs as a robust visual readout of early PDGFR signaling, we have identified several contradictory elements in the widely accepted model of PDGF activity. Employing CRISPR/Cas9 gene editing to disrupt the Pdgfra gene in two different murine cell lines, we show that in addition to the widely accepted function for PDGFR-beta in CDR formation, PDGFR-alpha is also clearly capable of eliciting CDRs. Moreover, we demonstrate activity for heterodimeric PDGF-AB ligand in the vigorous activation of PDGFR-beta homodimers to produce CDRs. These findings are key to a more complete understanding of PDGF ligand-receptor interactions and their downstream signaling consequences. This knowledge will allow for more rigorous experimental design in future studies of PDGFR signaling and its contributions to development and disease.


Asunto(s)
Becaplermina/metabolismo , Fibroblastos/metabolismo , Melanoma/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Sistemas CRISPR-Cas , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Ligandos , Melanoma/genética , Ratones , Unión Proteica , Multimerización de Proteína , Transducción de Señal
19.
Mol Biol Cell ; 31(16): 1774-1787, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-31967947

RESUMEN

Mechanical stimulation of fibroblasts induces changes in the actin cytoskeleton including stress fiber (SF) reinforcement and realignment. Here we characterize the nuclear response to mechanical stimulation (uniaxial cyclic stretch). Using fluorescence microscopy and quantitative image analysis we find that stretch-induced nuclear elongation and alignment perpendicular to the stretch vector are dependent on formin-regulated actin polymerization. The mechanosensitive transcription factors Yes-associated protein/Transcriptional coactivator with PDZ domain (YAP/TAZ) and myocardin-related transcription factor (MRTF-A, also known as MKL1 and MAL1) accumulate in the nucleus and activate their target genes in response to uniaxial cyclic stretch. We show that transmembrane actin nuclear (TAN) lines are induced by stretch stimulation and nuclear envelope (NE) proteins including nesprins, SUN2, and lamins form Linkers of the Nucleoskeleton and Cytoskeleton (LINC) complexes aligned with actin SFs. These NE structures are altered by pharmacological treatments (Cytochalasin D and Jasplakinolide) or genetic disruption (zyxin gene deletion) that alter actin, and their persistence requires maintenance of stretch stimulation. Nuclear pore complexes (NPCs) accumulate at TAN lines providing a potential mechanism for linking mechanical cues to NPC function.


Asunto(s)
Mecanorreceptores/metabolismo , Poro Nuclear/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Núcleo Celular/metabolismo , Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Proteínas Nucleares/metabolismo , Cultivo Primario de Células , Fibras de Estrés/metabolismo , Estrés Mecánico , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteínas Señalizadoras YAP
20.
Dev Cell ; 55(4): 468-482.e7, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-33058779

RESUMEN

Mechanical signals transmitted through the cytoplasmic actin cytoskeleton must be relayed to the nucleus to control gene expression. LIM domains are protein-protein interaction modules found in cytoskeletal proteins and transcriptional regulators. Here, we identify three LIM protein families (zyxin, paxillin, and FHL) whose members preferentially localize to the actin cytoskeleton in mechanically stimulated cells through their tandem LIM domains. A minimal actin-myosin reconstitution system reveals that representatives of all three families directly bind F-actin only in the presence of mechanical force. Point mutations at a site conserved in each LIM domain of these proteins disrupt tensed F-actin binding in vitro and cytoskeletal localization in cells, demonstrating a common, avidity-based mechanism. Finally, we find that binding to tensed F-actin in the cytoplasm excludes the cancer-associated transcriptional co-activator FHL2 from the nucleus in stiff microenvironments. This establishes direct force-activated F-actin binding as a mechanosensing mechanism by which cytoskeletal tension can govern nuclear localization.


Asunto(s)
Actinas/metabolismo , Proteínas con Dominio LIM/metabolismo , Mecanotransducción Celular , Citoesqueleto de Actina/metabolismo , Animales , Fenómenos Biomecánicos , Núcleo Celular/metabolismo , Secuencia Conservada , Adhesiones Focales/metabolismo , Humanos , Ratones , Fenilalanina/metabolismo , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA