Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
FASEB J ; 37(8): e23097, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37440280

RESUMEN

Colorectal cancer (CRC) is a high prevalence adenocarcinoma with progressive increases in metastasis-related mortality, but the mechanisms governing the extracellular matrix (ECM) degradation important for metastasis in CRC are not well-defined. We investigated a functional relationship between vimentin (Vim) and myosin 10 (Myo10), and whether this relationship is associated with cancer progression. We tested the hypothesis that Vim regulates the aggregation of Myo10 at the tips of cell extensions, which increases membrane-type 1 matrix metalloproteinase (MT1-MMP)-associated local collagen proteolysis and ECM degradation. Analysis of CRC samples revealed colocalization of Vim with Myo10 and MT1-MMP in cell extensions adjacent to sites of collagen degradation, suggesting an association with local cell invasion. We analyzed cultured CRC cells and fibroblasts and found that Vim accelerates aggregation of Myo10 at cell tips, which increases the cell extension rate. Vim stabilizes the interaction of Myo10 with MT1-MMP, which in turn increases collagenolysis. Vim depletion reduced the aggregation of Myo10 at the cell extension tips and MT1-MMP-dependent collagenolysis. We propose that Vim interacts with Myo10, which in turn associates with MT1-MMP to facilitate the transport of these molecules to the termini of cell extensions and there enhance cancer invasion of soft connective tissues.


Asunto(s)
Neoplasias Colorrectales , Metaloproteinasa 14 de la Matriz , Humanos , Metaloproteinasa 14 de la Matriz/metabolismo , Vimentina/metabolismo , Colágeno , Miosinas
2.
J Cell Physiol ; 237(5): 2451-2468, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35150133

RESUMEN

Tissue fibrosis manifests as excessive deposition of compacted, highly aligned collagen fibrils, which interfere with organ structure and function. Cells in collagen-rich lesions often exhibit marked overexpression of discoidin domain receptor 1 (DDR1), which is linked to increased collagen compaction through the association of DDR1 with the Ca2+ -dependent nonmuscle myosin IIA (NMIIA). We examined the functional relationship between DDR1 and the transient receptor potential vanilloid type 4 (TRPV4) channel, a Ca2+ -permeable ion channel that is implicated in collagen compaction. Fibroblasts expressing high levels of DDR1 were used to model cells in lesions with collagen compaction. In these cells, the expression of the ß1 integrin was deleted to simplify studies of DDR1 function. Compared with DDR1 wild-type cells, high DDR1 expression was associated with increased Ca2+ influx through TRPV4, enrichment of TRPV4 in collagen adhesions, and enhanced contractile activity mediated by NMIIA. At cell adhesion sites to collagen, DDR1 associated with TRPV4, which enhanced DDR1-mediated collagen alignment and compaction. We conclude that DDR1 regulates Ca2+ influx through the TRPV4 channel to promote critical, DDR1-mediated processes that are important in lesions with collagen compaction and alignment.


Asunto(s)
Calcio , Receptor con Dominio Discoidina 1 , Calcio/metabolismo , Calcio de la Dieta , Uniones Célula-Matriz/metabolismo , Colágeno/metabolismo , Receptor con Dominio Discoidina 1/genética , Miosinas/metabolismo , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo
3.
J Cell Sci ; 134(6)2021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-33558312

RESUMEN

Vimentin is a structural protein that is required for mesenchymal cell migration and directly interacts with actin, ß1 integrin and paxillin. We examined how these interactions enable vimentin to regulate cell migration on collagen. In fibroblasts, depletion of vimentin increased talin-dependent activation of ß1 integrin by more than 2-fold. Loss of vimentin was associated with reduction of ß1 integrin clustering by 50% and inhibition of paxillin recruitment to focal adhesions by more than 60%, which was restored by vimentin expression. This reduction of paxillin was associated with 65% lower Cdc42 activation, a 60% reduction of cell extension formation and a greater than 35% decrease in cell migration on collagen. The activation of PAK1, a downstream effector of Cdc42, was required for vimentin phosphorylation and filament maturation. We propose that vimentin tunes cell migration through collagen by acting as an adaptor protein for focal adhesion proteins, thereby regulating ß1 integrin activation, resulting in well-organized, mature integrin clusters.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Colágeno , Integrina beta1 , Adhesión Celular , Movimiento Celular , Análisis por Conglomerados , Integrina beta1/genética , Integrina beta1/metabolismo , Paxillin/genética , Paxillin/metabolismo , Vimentina/genética , Vimentina/metabolismo
4.
J Cell Sci ; 130(13): 2196-2208, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28526784

RESUMEN

Fibroblasts remodel extracellular matrix collagen, in part, through phagocytosis. This process requires formation of cell extensions, which in turn involves interaction of the actin-binding protein flightless-1 (FliI) with non-muscle myosin IIA (NMMIIA; heavy chain encoded by MYH9) at cell-matrix adhesion sites. As Ca2+ plays a central role in controlling actomyosin-dependent functions, we examined how Ca2+ controls the generation of cell extensions and collagen remodeling. Ratio fluorimetry demonstrated localized Ca2+ influx at the extensions of fibroblasts. Western blotting and quantitative (q)PCR showed high expression levels of the Ca2+-permeable transient receptor potential vanilloid-4 (TRPV4) channel, which co-immunoprecipitated with ß1 integrin and localized to adhesions. Treatment with α2ß1-integrin-blocking antibody or the TRPV4-specific antagonist AB159908, as well as reduction of TRPV4 expression through means of siRNA, blocked Ca2+ influx. These treatments also inhibited the interaction of FliI with NMMIIA, reduced the number and length of cell extensions, and blocked collagen remodeling. Pulldown assays showed that Ca2+ depletion inhibited the interaction of purified FliI with NMMIIA filaments. Fluorescence resonance energy transfer experiments showed that FliI-NMMIIA interactions require Ca2+ influx. We conclude that Ca2+ influx through the TRPV4 channel regulates FliI-NMMIIA interaction, which in turn enables generation of the cell extensions essential for collagen remodeling.


Asunto(s)
Señalización del Calcio/genética , Proteínas Portadoras/genética , Colágeno/genética , Miosina Tipo IIA no Muscular/genética , Canales Catiónicos TRPV/genética , Animales , Proteínas Portadoras/metabolismo , Uniones Célula-Matriz/genética , Colágeno/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Humanos , Integrina beta1/genética , Ratones , Proteínas de Microfilamentos , Cadenas Pesadas de Miosina , Células 3T3 NIH , Miosina Tipo IIA no Muscular/metabolismo , Fagocitosis , Mapas de Interacción de Proteínas/genética , ARN Interferente Pequeño/genética , Canales Catiónicos TRPV/metabolismo , Transactivadores
5.
Cell Rep ; 18(7): 1774-1790, 2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28199848

RESUMEN

Discoidin domain receptor 1 (DDR1) is a tyrosine kinase collagen adhesion receptor that mediates cell migration through association with non-muscle myosin IIA (NMIIA). Because DDR1 is implicated in cancer fibrosis, we hypothesized that DDR1 interacts with NMIIA to enable collagen compaction by traction forces. Mechanical splinting of rat dermal wounds increased DDR1 expression and collagen alignment. In periodontal ligament of DDR1 knockout mice, collagen mechanical reorganization was reduced >30%. Similarly, cultured cells with DDR1 knockdown or expressing kinase-deficient DDR1d showed 50% reduction of aligned collagen. Tractional remodeling of collagen was dependent on DDR1 clustering, activation, and interaction of the DDR1 C-terminal kinase domain with NMIIA filaments. Collagen remodeling by traction forces, DDR1 tyrosine phosphorylation, and myosin light chain phosphorylation were increased on stiff versus soft substrates. Thus, DDR1 clustering, activation, and interaction with NMIIA filaments enhance the collagen tractional remodeling that is important for collagen compaction in fibrosis.


Asunto(s)
Colágeno/metabolismo , Receptor con Dominio Discoidina 1/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Animales , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Femenino , Fibrosis/metabolismo , Ratones , Ratones Noqueados , Cadenas Ligeras de Miosina/metabolismo , Fosforilación/fisiología , Ratas , Ratas Wistar , Proteínas Tirosina Quinasas Receptoras/metabolismo
6.
Mol Biol Cell ; 26(12): 2279-97, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25877872

RESUMEN

We examined the role of the actin-capping protein flightless I (FliI) in collagen remodeling by mouse fibroblasts. FliI-overexpressing cells exhibited reduced spreading on collagen but formed elongated protrusions that stained for myosin10 and fascin and penetrated pores of collagen-coated membranes. Inhibition of Cdc42 blocked formation of cell protrusions. In FliI-knockdown cells, transfection with constitutively active Cdc42 did not enable protrusion formation. FliI-overexpressing cells displayed increased uptake and degradation of exogenous collagen and strongly compacted collagen fibrils, which was blocked by blebbistatin. Mass spectrometry analysis of FliI immunoprecipitates showed that FliI associated with nonmuscle myosin IIA (NMMIIA), which was confirmed by immunoprecipitation. GFP-FliI colocalized with NMMIIA at cell protrusions. Purified FliI containing gelsolin-like domains (GLDs) 1-6 capped actin filaments efficiently, whereas FliI GLD 2-6 did not. Binding assays showed strong interaction of purified FliI protein (GLD 1-6) with the rod domain of NMMIIA (kD = 0.146 µM), whereas FliI GLD 2-6 showed lower binding affinity (kD = 0.8584 µM). Cells expressing FliI GLD 2-6 exhibited fewer cell extensions, did not colocalize with NMMIIA, and showed reduced collagen uptake compared with cells expressing FliI GLD 1-6. We conclude that FliI interacts with NMMIIA to promote cell extension formation, which enables collagen remodeling in fibroblasts.


Asunto(s)
Extensiones de la Superficie Celular/metabolismo , Colágeno/metabolismo , Proteínas del Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Miosina Tipo IIA no Muscular/metabolismo , Animales , Proteínas Portadoras , Humanos , Ratones , Proteínas de Microfilamentos , Cadenas Pesadas de Miosina , Unión Proteica , Transactivadores
7.
Biochim Biophys Acta ; 1853(7): 1715-24, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25871908

RESUMEN

Cell fusion process is a critical, rate-limiting step in osteoclastogenesis but the mechanisms that regulate fusopod formation are not defined. We characterized fusopod generation in cultured pre-osteoclasts derived from cells stably transfected with a plasmid that expressed a short, actin filament binding peptide (Lifeact) fused to mEGFP that enables localization of actin filaments in living cells. Fusion was initiated at fusopods, which are cell extensions of width >2 µm and that are immunostained for myosin-X at the extension tips. Fusopods formed at the leading edge of larger migrating cells and from the tail of adjacent smaller cells, both of which migrated in the same direction. Staining for DC-STAMP was circumferential and did not localize to cell-cell fusion sites. Compared with wild-type cells, monocytes null for Rac1 exhibited 6-fold fewer fusopods and formed 4-fold fewer multinucleated osteoclasts. From time-lapse images we found that fusion was temporally related to the formation of coherent and spatially isolated bands of actin filaments that originated in cell bodies and extended into the fusopods. These bands of actin filaments were involved in cell fusion after approaching cells formed initial contacts. We conclude that the formation of fusopods is regulated by Rac1 to initiate intercellular contact during osteoclastogenesis. This step is followed by the tightly regulated assembly of bands of actin filaments in fusopods, which lead to closure of the intercellular gap and finally, cell fusion. These novel, actin-dependent processes are important for fusion processes in osteoclastogenesis.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Fusión Celular , Osteoclastos/citología , Osteoclastos/metabolismo , Osteogénesis , Citoesqueleto de Actina/efectos de los fármacos , Actinas/metabolismo , Animales , Técnicas de Cultivo de Célula , Línea Celular , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Ligando RANK/farmacología , Solubilidad , Proteína de Unión al GTP rac1/metabolismo
8.
J R Soc Interface ; 12(102): 20141074, 2015 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-25392399

RESUMEN

The mechanical properties of extracellular matrix proteins strongly influence cell-induced tension in the matrix, which in turn influences cell function. Despite progress on the impact of elastic behaviour of matrix proteins on cell-matrix interactions, little is known about the influence of inelastic behaviour, especially at the large and slow deformations that characterize cell-induced matrix remodelling. We found that collagen matrices exhibit deformation rate-dependent behaviour, which leads to a transition from pronounced elastic behaviour at fast deformations to substantially inelastic behaviour at slow deformations (1 µm min(-1), similar to cell-mediated deformation). With slow deformations, the inelastic behaviour of floating gels was sensitive to collagen concentration, whereas attached gels exhibited similar inelastic behaviour independent of collagen concentration. The presence of an underlying rigid support had a similar effect on cell-matrix interactions: cell-induced deformation and remodelling were similar on 1 or 3 mg ml(-1) attached collagen gels while deformations were two- to fourfold smaller in floating gels of high compared with low collagen concentration. In cross-linked collagen matrices, which did not exhibit inelastic behaviour, cells did not respond to the presence of the underlying rigid foundation. These data indicate that at the slow rates of collagen compaction generated by fibroblasts, the inelastic responses of collagen gels, which are influenced by collagen concentration and the presence of an underlying rigid foundation, are important determinants of cell-matrix interactions and mechanosensation.


Asunto(s)
Colágeno/química , Reactivos de Enlaces Cruzados/química , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Células 3T3 , Animales , Bovinos , Adhesión Celular , Comunicación Celular , Células Cultivadas , Elasticidad , Análisis de Fourier , Geles , Ratones , Microscopía Confocal , Microscopía Fluorescente , Estrés Mecánico
9.
J Cell Physiol ; 230(2): 327-36, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24962729

RESUMEN

The adhesion of cardiac fibroblasts to the glycated collagen interstitium in diabetics is associated with de novo expression of the α11 integrin, myofibroblast formation and cardiac fibrosis. We examined how methylglyoxal-glycated collagen regulates α11 integrin expression. In cardiac fibroblasts plated on glycated collagen but not glycated fibronectin, there was markedly increased α11 integrin and α-smooth muscle actin expression. Compared with native collagen, binding of purified α11ß1 integrin to glycated collagen was reduced by >fourfold, which was consistent with reduced fibroblast attachment to glycated collagen. Glycated collagen strongly enhanced the expression of TGF-ß2 but not TGF-ß1 or TGF-ß3. The increased expression of TGF-ß2 was inhibited by triple helical collagen peptides that mimic the α11ß1 integrin binding site on type I collagen. In cardiac fibroblasts transfected with α11 integrin luciferase promoter constructs, glycated collagen activated the α11 integrin promoter. Analysis of α11 integrin promoter truncation mutants showed a novel Smad2/3 binding site located between -809 and -1300 nt that was required for promoter activation. We conclude that glycated collagen in the cardiac interstitium triggers an autocrine TGF-ß2 signaling pathway that stimulates α11 integrin expression through Smad2/3 binding elements in the α11 integrin promoter, which is important for myofibroblast formation and fibrosis.


Asunto(s)
Colágeno/metabolismo , Fibronectinas/metabolismo , Integrinas/metabolismo , Miofibroblastos/metabolismo , Receptores de Colágeno/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Células Cultivadas , Productos Finales de Glicación Avanzada , Humanos , Proteína Smad2/metabolismo
10.
Periodontol 2000 ; 63(1): 48-58, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23931053

RESUMEN

Adhesion molecules expressed by periodontal connective tissue cells are involved in cell migration, matrix remodeling and inflammatory responses to infection. Currently, the processes by which the biologic activity of these molecules are appropriately regulated in time and space to preserve tissue homeostasis, and to control inflammatory responses and tissue regeneration, are not defined. As cell adhesions are heterogeneous, dynamic, contain a complex group of interacting molecules and are strongly influenced by the type of substrate to which they adhere, we focus on how cell adhesions in periodontal connective tissues contribute to information generation and processing that regulate periodontal structure and function. We also consider how proteomic methods can be applied to discover novel cell-adhesion proteins that could potentially contribute to the form and function of periodontal tissues.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Periodoncio/fisiología , Proteómica , Adhesión Celular/fisiología , Comunicación Celular/fisiología , Movimiento Celular/fisiología , Células del Tejido Conectivo/fisiología , Matriz Extracelular/fisiología , Homeostasis/fisiología , Humanos , Inflamación , Mediadores de Inflamación/inmunología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Regeneración/fisiología
11.
Cardiovasc Res ; 96(2): 265-75, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22869616

RESUMEN

AIMS: Diabetic cardiomyopathy is characterized by the production of a disorganized fibrotic matrix in the absence of coronary atherosclerosis and hypertension. We examined whether adhesion of cardiac fibroblasts to glycated collagens mediates the differentiation of pro-fibrotic myofibroblasts, which may contribute to cardiac fibrosis. METHODS AND RESULTS: By microarray, we found that methylglyoxal-treated collagen selectively enhanced α11 integrin expression in human cardiac fibroblasts, while levels of other collagen-binding integrins (α1, α2, and α10) were unchanged. Similar increases in α11 integrin mRNA and protein expression were observed in cardiac fibroblasts from streptozotocin (STZ)-treated Sprague-Dawley rats. In human cardiac fibroblasts plated on methyglyoxal-treated collagen and in cardiac fibroblasts from diabetic rats, transforming growth factor (TGF)-ß2 but not TGF-ß1 or TGF-ß3 was increased compared with controls. Knock-down of α11 integrin and TGF-ß receptors with small-interfering RNA blocked the increased expression of TGF-ß2, α-smooth muscle actin (α-SMA), and α11 integrin that were induced in cells plated on methylglyoxal-treated collagen. Further, inhibition of Smad3 signalling blocked methylglyoxal-collagen up-regulation of α11 integrin and α-SMA expression. Rats with STZ-induced diabetes exhibited increased phosphorylation of Smad3 in cardiac tissues compared with control rats. CONCLUSION: Interactions between α11 integrins and the Smad-dependent TGF-ß2 signalling may contribute to the formation of pro-fibrotic myofibroblasts and the development of a fibrotic interstitium in diabetic cardiomyopathy.


Asunto(s)
Diferenciación Celular , Cardiomiopatías Diabéticas/patología , Cadenas alfa de Integrinas/metabolismo , Miocardio/patología , Miofibroblastos/patología , Actinas/metabolismo , Animales , Adhesión Celular , Células Cultivadas , Colágeno/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Cardiomiopatías Diabéticas/metabolismo , Humanos , Masculino , Piruvaldehído , Ratas , Ratas Sprague-Dawley , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo
12.
FASEB J ; 26(8): 3260-72, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22581781

RESUMEN

The role of adhesion-associated actin-binding proteins in cell migration is not well defined. In mouse fibroblasts we screened for focal adhesion-associated proteins that were isolated with collagen-coated beads and detected by tandem mass spectrometry. We identified flightless I (FliI) as an actin-binding protein in focal adhesion fractions, which was verified by immunoblotting. By confocal microscopy most FliI was distributed throughout the cytosol and in focal adhesions. By sedimentation assays and in vitro binding assays, we found that FliI associates with actin filaments and actin monomers. Assays using purified proteins showed that FliI inhibits actin polymerization and caps but does not sever actin filaments. Cells with FliI knockdown or cells overexpressing FliI migrated more or less rapidly, respectively, than wild-type controls. Compared with controls, cells with FliI knockdown were less adherent than wild-type cells, exhibited reduced numbers of focal adhesions containing activated ß1 integrins and vinculin, and exhibited increased incorporation of actin monomers into nascent filaments at focal adhesions. These data indicate that FliI regulates cell migration through its localization to focal adhesions and its ability to cap actin filaments, which collectively affect focal adhesion maturation.


Asunto(s)
Proteínas de Capping de la Actina/metabolismo , Proteínas del Citoesqueleto/fisiología , Adhesiones Focales/metabolismo , Actinas/fisiología , Animales , Proteínas Portadoras , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Ratones , Proteínas de Microfilamentos , Transactivadores
13.
Biochem J ; 445(1): 57-67, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22489840

RESUMEN

Cells in mechanically challenged environments cope with high-amplitude exogenous forces that can lead to cell death, but the mechanisms that mediate force-induced apoptosis and the identity of mechanoprotective cellular factors are not defined. We assessed apoptosis in NIH 3T3 and HEK (human embryonic kidney)-293 cells exposed to tensile forces applied through ß1-integrins. Apoptosis was mediated by Rac-dependent activation of p38α. Depletion of Pak1 (p21-activated kinase 1), a downstream effector of Rac, prevented force-induced p38 activation and apoptosis. Rac was recruited to sites of force transfer by filamin A, which inhibited force-induced apoptosis mediated by Rac and p38α. We conclude that, in response to tensile force, filamin A regulates Rac-dependent signals, which induce apoptosis through Pak1 and p38.


Asunto(s)
Apoptosis , Proteínas Contráctiles/metabolismo , Mecanotransducción Celular , Proteínas de Microfilamentos/metabolismo , Transducción de Señal , Quinasas p21 Activadas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Caspasa 3/metabolismo , Células Cultivadas , Filaminas , Humanos , Immunoblotting , Riñón/citología , Riñón/metabolismo , Ratones , Células 3T3 NIH , ARN Interferente Pequeño/genética , Quinasas p21 Activadas/antagonistas & inhibidores , Quinasas p21 Activadas/genética
14.
J Cell Biol ; 197(1): 115-29, 2012 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-22472442

RESUMEN

Leukocytes arrested on inflamed endothelium via integrins are subjected to force imparted by flowing blood. How leukocytes respond to this force and resist detachment is poorly understood. Live-cell imaging with Lifeact-transfected U937 cells revealed that force triggers actin polymerization at upstream α4ß1 integrin adhesion sites and the adjacent cortical cytoskeleton. Scanning electron microscopy revealed that this culminates in the formation of structures that anchor monocyte adhesion. Inhibition of actin polymerization resulted in cell deformation, displacement, and detachment. Transfection of dominant-negative constructs and inhibition of function or expression revealed key signaling steps required for upstream actin polymerization and adhesion stabilization. These included activation of Rap1, phosphoinositide 3-kinase γ isoform, and Rac but not Cdc42. Thus, rapid signaling and structural adaptations enable leukocytes to stabilize adhesion and resist detachment forces.


Asunto(s)
Actinas/metabolismo , Integrina alfa4beta1/metabolismo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Adhesión Celular , Células Cultivadas , Humanos , Células U937
15.
J Biol Chem ; 286(39): 34184-98, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21828045

RESUMEN

The formation of adhesion complexes is the rate-limiting step for collagen phagocytosis by fibroblasts, but the role of Ca(2+) and the potential interactions of actin-binding proteins in regulating collagen phagocytosis are not well defined. We found that the binding of collagen beads to fibroblasts was temporally and spatially associated with actin assembly at nascent phagosomes, which was absent in gelsolin null cells. Analysis of tryptic digests isolated from gelsolin immunoprecipitates indicated that non-muscle (NM) myosin IIA may bind to gelsolin. Immunostaining and immunoprecipitation showed that gelsolin and NM myosin IIA associated at collagen adhesion sites. Gelsolin and NM myosin IIA were both required for collagen binding and internalization. Collagen binding to cells initiated a prolonged increase of [Ca(2+)](i), which was absent in cells null for gelsolin or NM myosin IIA. Collagen bead-induced increases of [Ca(2+)](i) were associated with phosphorylation of the myosin light chain, which was dependent on gelsolin. NM myosin IIA filament assembly, which was dependent on myosin light chain phosphorylation and increased [Ca(2+)](i), also required gelsolin. Ionomycin-induced increases of [Ca(2+)](i) overcame the block of myosin filament assembly in gelsolin null cells. We conclude that gelsolin and NM myosin IIA interact at collagen adhesion sites to enable NM myosin IIA filament assembly and localized, Ca(2+)-dependent remodeling of actin at the nascent phagosome and that these steps are required for collagen phagocytosis.


Asunto(s)
Calcio/metabolismo , Colágeno/metabolismo , Fibroblastos/metabolismo , Gelsolina/metabolismo , Miosina Tipo IIA no Muscular/metabolismo , Fagocitosis/fisiología , Animales , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Células Cultivadas , Colágeno/genética , Fibroblastos/citología , Gelsolina/genética , Humanos , Ionomicina/farmacología , Ionóforos/farmacología , Ratones , Ratones Noqueados , Miosina Tipo IIA no Muscular/genética , Fagocitosis/efectos de los fármacos , Fagosomas/genética , Fagosomas/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/fisiología
16.
J Cell Sci ; 122(Pt 15): 2769-81, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19596799

RESUMEN

During the development of pressure-induced cardiac hypertrophy, fibroblasts are activated to become myofibroblasts, which exhibit actin-cytoskeletal remodeling and express alpha-smooth muscle actin (SMA; encoded by ACTA2). Currently, the mechanosensing signaling pathways that regulate SMA expression are not defined. Because focal-adhesion complexes are putative mechanosensing organelles, we examined the role of focal adhesion kinase (FAK) and its interaction with gelsolin in the regulation of SMA expression. We subjected NIH3T3 cells to tensile forces (0.65 pN/mum(2)) by using collagen-coated magnetite beads attached to integrins. After stimulation by mechanical force, FAK and gelsolin were recruited to magnetite beads and there was increased phosphorylation of Tyr397FAK. Mechanical force enhanced SMA promoter activity by twofold; this increased activity was blocked by FAK knockdown using siRNA and by deletion of gelsolin. Force-induced nuclear translocation of MRTF-A, a transcriptional co-activator of SMA that is regulated by actin filaments, was also reduced by FAK knockdown. Phosphatidylinositol (4,5)-bisphosphate [PtdIns(4,5)P(2)], which uncaps gelsolin from actin filaments, was enriched at sites of force application. Type-I phosphatidylinositol 4-phosphate 5 kinase-gamma (PIP5KIgamma), which generates PtdIns(4,5)P(2), associated with FAK and was required for force-mediated SMA-promoter activity and actin assembly. Catalytically inactive PIP5KIgamma inhibited force-induced phosphorylation of FAK at Tyr397. These data suggest a novel pathway in which mechanosensing by FAK regulates actin assembly via gelsolin and the activity of PIP5KIgamma; actin assembly in turn controls SMA expression via MRTF-A.


Asunto(s)
Actinas/genética , Quinasa 1 de Adhesión Focal/metabolismo , Gelsolina/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Estrés Mecánico , Actinas/metabolismo , Animales , Técnica del Anticuerpo Fluorescente , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/genética , Gelsolina/genética , Ratones , Células 3T3 NIH , Fosfatos de Fosfatidilinositol/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Regiones Promotoras Genéticas , ARN Interferente Pequeño/farmacología , Transactivadores/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...