Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Cell Physiol ; 236(2): 1270-1280, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32643295

RESUMO

Many adult connective tissues undergo continuous remodeling to maintain matrix homeostasis. Physiological remodeling involves the degradation of collagen fibers by the intracellular cathepsin-dependent phagocytic pathway. We considered that a multidomain, small GTPase activating protein, IQGAP1, which is involved in the generation of cell extensions, is required for collagen phagocytosis, possibly arising from its interactions with cdc42 and the actin-binding protein Flightless I (FliI). We examined the role of IQGAP1 in collagen phagocytosis by human gingival fibroblasts (HGFs) and by IQGAP1+/+ and IQGAP1-/- mouse embryonic fibroblasts. IQGAP1 was strongly expressed by HGFs, localized to vinculin-stained cell adhesions and sites where cell extensions are initiated, and colocalized with FliI. Immunoprecipitation showed that IQGAP1 associated with FliI. HGFs showed 10-fold increases of collagen binding, 6-fold higher internalization, and 3-fold higher ß1 integrin activation between 30 and 180 min after incubation with collagen. Compared with IQGAP1+/+ fibroblasts, deletion of IQGAP1 reduced collagen binding (1.4-fold), collagen internalization (3-fold), ß1 integrin activation (2-fold), and collagen degradation (1.8-fold). We conclude that IQGAP1 affects collagen remodeling through its regulation of phagocytic degradation pathways, which may involve the interaction of IQGAP1 with FliI.


Assuntos
Colágeno/genética , Proteínas dos Microfilamentos/genética , Fagocitose/genética , Transativadores/genética , Proteína cdc42 de Ligação ao GTP/genética , Proteínas Ativadoras de ras GTPase/genética , Animais , Adesão Celular/genética , Colágeno/metabolismo , Fibroblastos/metabolismo , Gengiva/metabolismo , Gengiva/patologia , Humanos , Integrina beta1 , Camundongos , Proteínas Monoméricas de Ligação ao GTP/genética , Ligação Proteica/genética , Transdução de Sinais/genética
2.
Med Res Rev ; 32(5): 999-1025, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22886630

RESUMO

Gelsolin, a Ca(2+) -regulated actin filament severing, capping, and nucleating protein, is an ubiquitous, multifunctional regulator of cell structure and metabolism. More recent data show that gelsolin can act as a transcriptional cofactor in signal transduction and its own expression and function can be influenced by epigenetic changes. Here, we review the functions of the plasma and cytoplasmic forms of gelsolin, and their manifold impacts on cancer, apoptosis, infection and inflammation, cardiac injury, pulmonary diseases, and aging. An improved understanding of the functions and regulatory mechanisms of gelsolin may lead to new considerations of this protein as a potential biomarker and/or therapeutic target.


Assuntos
Doença , Gelsolina/metabolismo , Saúde , Animais , Apoptose , Gelsolina/química , Gelsolina/genética , Humanos , Fagocitose , Transdução de Sinais
3.
Mol Biol Cell ; 20(5): 1269-79, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19144823

RESUMO

Cells in mechanically active environments are subjected to high-amplitude exogenous forces that can lead to cell death. Filamin A (FLNa) may protect cells from mechanically induced death by mechanisms that are not yet defined. We found that mechanical forces applied through integrins enhanced Rac-mediated lamellae formation in FLNa-null but not FLNa-expressing cells. Suppression of force-induced lamella formation was mediated by repeat 23 of FLNa, which also binds FilGAP, a recently discovered Rac GTPase-activating protein (GAP). We found that FilGAP is targeted to sites of force transfer by FLNa. This force-induced redistribution of FilGAP was essential for the suppression of Rac activity and lamellae formation in cells treated with tensile forces. Depletion of FilGAP by small interfering RNA, inhibition of FilGAP activity by dominant-negative mutation or deletion of its FLNa-binding domain, all resulted in a dramatic force-induced increase of the percentage of annexin-V-positive cells. FilGAP therefore plays a role in protecting cells against force-induced apoptosis, and this function is mediated by FLNa.


Assuntos
Proteínas Contráteis/fisiologia , Proteínas Ativadoras de GTPase/fisiologia , Mecanotransdução Celular/fisiologia , Proteínas dos Microfilamentos/fisiologia , Citoesqueleto de Actina/metabolismo , Animais , Anexina A5/metabolismo , Apoptose , Sítios de Ligação , Linhagem Celular , Forma Celular , Proteínas Contráteis/genética , Proteínas Contráteis/metabolismo , Filaminas , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Humanos , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Transporte Proteico , Estresse Fisiológico , Proteínas rac de Ligação ao GTP/metabolismo
4.
Mol Biol Cell ; 19(12): 5032-46, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18799623

RESUMO

Rap1 enhances integrin-mediated adhesion but the link between Rap1 activation and integrin function in collagen phagocytosis is not defined. Mass spectrometry of Rap1 immunoprecipitates showed that the association of Rap1 with nonmuscle myosin heavy-chain II-A (NMHC II-A) was enhanced by cell attachment to collagen beads. Rap1 colocalized with NM II-A at collagen bead-binding sites. There was a transient increase in myosin light-chain phosphorylation after collagen-bead binding that was dependent on myosin light-chain kinase but not Rho kinase. Inhibition of myosin light-chain phosphorylation, but not myosin II-A motor activity inhibited collagen-bead binding and Rap activation. In vitro binding assays demonstrated binding of Rap1A to filamentous myosin rods, and in situ staining of permeabilized cells showed that NM II-A filaments colocalized with F-actin at collagen bead sites. Knockdown of NM II-A did not affect talin, actin, or beta1-integrin targeting to collagen beads but targeting of Rap1 and vinculin to collagen was inhibited. Conversely, knockdown of Rap1 did not affect localization of NM II-A to beads. We conclude that MLC phosphorylation in response to initial collagen-bead binding promotes NM II-A filament assembly; binding of Rap1 to myosin filaments enables Rap1-dependent integrin activation and enhanced collagen phagocytosis.


Assuntos
Colágeno/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Fagocitose/fisiologia , Proteínas rap1 de Ligação ao GTP/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Ativação Enzimática , Fibroblastos/citologia , Fibroblastos/fisiologia , Compostos Heterocíclicos de 4 ou mais Anéis/metabolismo , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Camundongos , Dados de Sequência Molecular , Quinase de Cadeia Leve de Miosina/genética , Quinase de Cadeia Leve de Miosina/metabolismo , Miosina não Muscular Tipo IIA/genética , Peptídeos/genética , Peptídeos/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Talina/genética , Talina/metabolismo , Proteínas rap1 de Ligação ao GTP/genética
5.
Am J Physiol Cell Physiol ; 293(3): C1049-58, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17615162

RESUMO

Cyclosporin A (CsA) inhibits collagen remodeling by interfering with the collagen-binding step of phagocytosis. In rapidly remodeling connective tissues such as human periodontium this interference manifests as marked tissue overgrowth and loss of function. Previous data have shown that CsA inhibits integrin-induced release of Ca(2+) from internal stores, which is required for the binding step of collagen phagocytosis. Because gelsolin is a Ca(2+)-dependent actin-severing protein that mediates collagen phagocytosis, we determined whether gelsolin is a CsA target. Compared with vehicle controls, CsA treatment of wild-type mice increased collagen accumulation by 60% in periodontal tissues; equivalent increases were seen in vehicle-treated gelsolin-null mice. Collagen degradation by phagocytosis in cultured gelsolin wild-type fibroblasts was blocked by CsA, comparable to levels of vehicle-treated gelsolin-null fibroblasts. In wild-type cells treated with CsA, collagen binding was similar to that of gelsolin-null fibroblasts transfected with a gelsolin-severing mutant and treated with vehicle. CsA blocked collagen-induced Ca(2+) fluxes subjacent to bound collagen beads, gelsolin recruitment, and actin assembly at bead sites. CsA reduced gelsolin-dependent severing of actin in wild-type cells to levels similar to those in gelsolin-null fibroblasts. We conclude that CsA-induced accumulation of collagen in the extracellular matrix involves disruption of the actin-severing properties of gelsolin, thereby inhibiting the binding step of collagen phagocytosis.


Assuntos
Colágeno/metabolismo , Ciclosporina/farmacologia , Gelsolina/metabolismo , Imunossupressores/farmacologia , Fagocitose/efeitos dos fármacos , Actinas/metabolismo , Animais , Cálcio/metabolismo , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Masculino , Camundongos , Camundongos Knockout
6.
Gene ; 379: 51-61, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16797880

RESUMO

Cell stressors such as physical forces can activate Sp1-dependent genes but the regulatory mechanisms are not defined. We determined if the stress-induced MAP kinase, p38, can phosphorylate Sp1 and thereby regulate the Sp1 target gene FLNA. We used Rat-2 cells and human gingival fibroblasts to examine stress-induced activation of an Sp1-dependent gene and SL2 cells, an Sp1-deficient model system, to facilitate interaction studies of transfected Sp1 with regulatory factors. Mechanical stress applied to Rat-2 cells increased promoter activity of the Sp1 target gene filamin A by >5-fold; activation was blocked by mutations to Sp1 binding sites in the filamin A promoter. Transfection experiments in SL2 cells with Sp1 expression vectors showed that when co-transfected with constitutively active p38, wild-type Sp1 but not an Sp1 binding mutant, increased promoter activity of the Sp1 target gene, filamin A, and enhanced binding of nuclear extracts to a filamin A promoter oligonucleotide. Filamin A promoter activity was blocked by dominant negative p38. Sp1 that was phosphorylated at Thr453 and Thr739 by constitutively active p38 bound to the filamin A promoter more effectively than un-phosphorylated Sp1. Recombinant active p38 phosphorylated wild-type Sp1 in vitro while the Sp1 Thr453Thr739 double mutant protein showed >3-fold reduction of phosphorylation. We conclude that stress activation of p38 phosphorylates Sp1 at specific threonine residues, modifications which in turn enhance the expression of Sp1-dependent genes.


Assuntos
Fator de Transcrição Sp1/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Animais , Sítios de Ligação , Células Cultivadas , Proteínas Contráteis/genética , Proteínas Contráteis/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Filaminas , Regulação da Expressão Gênica , Humanos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Ratos , Transdução de Sinais , Fator de Transcrição Sp1/genética , Estresse Mecânico , Treonina/genética , Treonina/metabolismo , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
Mol Biol Cell ; 16(12): 5514-27, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16176974

RESUMO

Cortactin regulates the strength of nascent N-cadherin-mediated intercellular adhesions through a tyrosine phosphorylation-dependent mechanism. Currently, the functional significance of cortactin phosphorylation and the kinases responsible for the regulation of adhesion strength are not defined. We show that the nonreceptor tyrosine kinase Fer phosphorylates cadherin-associated cortactin and that this process is involved in mediating intercellular adhesion strength. In wild-type fibroblasts N-cadherin ligation-induced transient phosphorylation of Fer, indicating that junction formation activates Fer kinase. Tyrosine phosphorylation of cortactin after N-cadherin ligation was strongly reduced in fibroblasts expressing only catalytically inactive Fer (D743R), compared with wild-type cells. In wild-type cells, N-cadherin-coated bead pull-off assays induced fourfold greater endogenous N-cadherin association than in D743R cells. Fluorescence recovery after photobleaching showed that GFP-N-cadherin mobility at nascent contacts was 50% faster in wild-type than D743R cells. In shear wash-off assays, nascent intercellular adhesion strength was twofold higher in wild-type than D743R cells. Cortactin recruitment to adhesions was independent of Fer kinase activity, but was impacted by N-cadherin ligation-provoked Rac activation. We conclude that N-cadherin ligation induces Rac-dependent cortactin recruitment and Fer-dependent cortactin phosphorylation, which in turn promotes enhanced mobilization and interaction of surface expressed N-cadherin in contacting cells.


Assuntos
Caderinas/fisiologia , Adesão Celular/fisiologia , Cortactina/metabolismo , Proteínas Tirosina Quinases/metabolismo , Animais , Linhagem Celular , Ativação Enzimática , GTP Fosfo-Hidrolases/metabolismo , Cinética , Camundongos , Fosforilação , Ratos
8.
J Biol Chem ; 280(24): 23103-13, 2005 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-15811857

RESUMO

Decorin is a small, leucine-rich proteoglycan that binds to collagen and regulates fibrillogenesis. We hypothesized that decorin binding to collagen inhibits phagocytosis of collagen fibrils. To determine the effects of decorin on collagen degradation, we analyzed phagocytosis of collagen and collagen/decorin-coated fluorescent beads by Rat-2 and gingival fibroblasts. Collagen beads bound to gingival cells by alpha2beta1 integrins. Binding and internalization of decorin/collagen-coated beads decreased dose-dependently with increasing decorin concentration (p < 0.001). Inhibition of binding was sustained over 5 h (p < 0.001) and was attributed to interactions between decorin and collagen and not to decorin-collagen receptor interactions. Both the non-glycosylated decorin core protein and the thermally denatured decorin significantly inhibited collagen bead binding (approximately 50 and 89%, respectively; p < 0.05). Mimetic peptides corresponding to leucine-rich repeats 1-3, encompassed by a collagen-binding approximately 11-kDa cyanogen bromide fragment of decorin and leucine-rich repeats 4 and 5, previously shown to bind to collagen, were tested for their ability to inhibit collagen bead binding. Although the synthetic peptide 3 alone exhibited saturable binding to collagen, neither peptides 3 nor 1 and 2 markedly inhibited phagocytosis. Leucine-rich repeat 3 bound to a triple helical peptide containing the alpha2 integrin-binding site of collagen. When collagen beads were co-incubated with peptides 3 and 4, inhibition of collagen phagocytosis (55%) was equivalent to intact native/recombinant core protein. Thus a novel collagen binding domain in decorin acts cooperatively with leucine-rich repeat 4 to mask the alpha2beta1 integrin-binding site on collagen, an important sequence for the phagocytosis of collagen fibrils.


Assuntos
Colágeno/química , Fibroblastos/metabolismo , Fagocitose , Proteoglicanas/fisiologia , Animais , Sítios de Ligação , Ligação Competitiva , Bovinos , Adesão Celular , Linhagem Celular , Membrana Celular/metabolismo , Movimento Celular , Dicroísmo Circular , Colágeno/metabolismo , Decorina , Relação Dose-Resposta a Droga , Proteínas da Matriz Extracelular , Citometria de Fluxo/métodos , Gengiva/citologia , Humanos , Integrina alfa2beta1/metabolismo , Cinética , Lisossomos/metabolismo , Espectrometria de Massas , Microscopia Confocal , Peptídeos/química , Ligação Proteica , Estrutura Terciária de Proteína , Proteoglicanas/metabolismo , Ratos , Proteínas Recombinantes/química , Fatores de Tempo
9.
J Biol Chem ; 280(8): 7273-84, 2005 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-15591055

RESUMO

The mitogen-activated protein kinase p38 is activated by mechanical force, but the cellular elements that mediate force-induced p38 phosphorylation are not defined. As alpha-smooth muscle actin (SMA) is an actin isoform associated with force generation in fibroblasts, we asked if SMA participates in the activation of p38 by force. Tensile forces (0.65 pn/mum(2)) generated by magnetic fields were applied to collagen-coated magnetite beads bound to Rat-2 cells. Immunoblotting showed that p38alpha was the predominant p38 isoform. Analysis of bead-associated proteins demonstrated that SMA enrichment of collagen receptor complexes required the alpha2beta1 integrin. SMA was present almost entirely as filaments. Swinholide depolymerized SMA filaments and blocked force-induced p38 phosphorylation and force-induced increases of SMA. Knockdown of SMA (70% reduction) using RNA interference did not affect beta-actin but inhibited force-induced p38 phosphorylation by 50%. Inhibition of Rho kinase blocked SMA filament assembly, force-induced increases of SMA, and force-induced p38 activation. Force application increased SMA content and enhanced the association of phosphorylated p38 with SMA filaments. Blockade of p38 phosphorylation by SB203586 abrogated force-induced increases of SMA. In cells transfected with SMA promoter-beta-galactosidase fusion constructs, co-transfection with constitutively active p38 or MKK6 increased SMA promoter activity by 2.5-3-fold. Dominant negative p38 blocked force-induced activation of the SMA promoter. In SMA negative cells, there was no force-induced p38 phosphorylation. We conclude that force-induced p38 phosphorylation is dependent on an SMA filament-dependent pathway that uses a feed-forward amplification loop to synergize force-induced SMA expression with p38 activation.


Assuntos
Actinas/fisiologia , Fenômenos Biomecânicos , Músculo Liso/química , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Linhagem Celular , Integrina alfa2beta1/metabolismo , Músculo Liso/citologia , Fosforilação , Ligação Proteica , Isoformas de Proteínas , Ratos , Receptores de Colágeno , Proteínas rho de Ligação ao GTP/metabolismo
10.
J Cell Sci ; 117(Pt 21): 5117-31, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15383621

RESUMO

The regulation of N-cadherin-mediated intercellular adhesion strength in fibroblasts is poorly characterized; this is due, in part, to a lack of available quantitative models. We used a recombinant N-cadherin chimeric protein and a Rat 2 fibroblast, donor-acceptor cell model, to study the importance of cortical actin filaments and cortactin in the strengthening of N-cadherin adhesions. In wash-off assays, cytochalasin D (1 microM) reduced intercellular adhesion by threefold, confirming the importance of cortical actin filaments in strengthening of N-cadherin-mediated adhesions. Cortactin, an actin filament binding protein, spatially colocalized to, and directly associated with, nascent N-cadherin adhesion complexes. Transfection of Rat-2 cells with cortactin-specific, RNAi oligonucleotides reduced cortactin protein by 85% and intercellular adhesion by twofold compared with controls (P<0.005) using the donor-acceptor model. Cells with reduced cortactin exhibited threefold less N-cadherin-mediated intercellular adhesion strength compared with controls in wash-off assays using N-cadherin-coated beads. Immunoprecipitation and immunoblotting showed that N-cadherin-associated cortactin was phosphorylated on tyrosine residue 421 after intercellular adhesion. While tyrosine phosphorylation of cortactin was not required for recruitment to N-cadherin adhesions it was necessary for cadherin-mediated intercellular adhesion strength. Thus cortactin, and phosphorylation of its tyrosine residues, are important for N-cadherin-mediated intercellular adhesion strength.


Assuntos
Caderinas/metabolismo , Proteínas dos Microfilamentos/química , Actinas/química , Actinas/metabolismo , Animais , Adesão Celular , Linhagem Celular , Sobrevivência Celular , Cortactina , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Citometria de Fluxo , Proteínas de Fluorescência Verde/metabolismo , Immunoblotting , Imunoprecipitação , Proteínas dos Microfilamentos/metabolismo , Microscopia de Fluorescência , Microscopia de Vídeo , Modelos Biológicos , Oligonucleotídeos/química , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Proteínas Recombinantes/química , Fatores de Tempo , Transfecção , Tirosina/química
11.
J Biol Chem ; 279(39): 41047-57, 2004 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-15247242

RESUMO

The regulation of adherens junction formation in cells of mesenchymal lineage is of critical importance in tumorigenesis but is poorly characterized. As actin filaments are crucial components of adherens junction assembly, we studied the role of gelsolin, a calcium-dependent, actin severing protein, in the formation of N-cadherin-mediated intercellular adhesions. With a homotypic, donor-acceptor cell model and plates or beads coated with recombinant N-cadherin-Fc chimeric protein, we found that gelsolin spatially co-localizes to, and is transiently associated with, cadherin adhesion complexes. Fibroblasts from gelsolin-null mice exhibited marked reductions in kinetics and strengthening of N-cadherin-dependent junctions when compared with wild-type cells. Experiments with lanthanum chloride (250 microm) showed that adhesion strength was dependent on entry of calcium ions subsequent to N-cadherin ligation. Cadherin-associated gelsolin severing activity was required for localized actin assembly as determined by rhodamine actin monomer incorporation onto actin barbed ends at intercellular adhesion sites. Scanning electron microscopy showed that gelsolin was an important determinant of actin filament architecture of adherens junctions at nascent N-cadherin-mediated contacts. These data indicate that increased actin barbed end generation by the severing activity of gelsolin associated with N-cadherin regulates intercellular adhesion strength.


Assuntos
Fibroblastos/citologia , Actinas/química , Actinas/metabolismo , Animais , Caderinas/química , Caderinas/metabolismo , Cálcio/metabolismo , Adesão Celular , Linhagem Celular , Linhagem da Célula , Células Cultivadas , Citoesqueleto/metabolismo , Regulação para Baixo , Fibroblastos/metabolismo , Citometria de Fluxo , Gelsolina/química , Humanos , Immunoblotting , Cinética , Lantânio/farmacologia , Magnetismo , Camundongos , Microscopia Eletrônica , Microscopia Eletrônica de Varredura , Microscopia de Fluorescência , Microscopia de Vídeo , Modelos Biológicos , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes/metabolismo , Fatores de Tempo , Transfecção
12.
Mol Biol Cell ; 15(2): 588-99, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14617805

RESUMO

The role of gelsolin, a calcium-dependent actin-severing protein, in mediating collagen phagocytosis, is not defined. We examined alpha 2 beta 1 integrin-mediated phagocytosis in fibroblasts from wild-type (WT) and gelsolin knockout (Gsn(-)) mice. After initial contact with collagen beads, collagen binding and internalization were 60% lower in Gsn(-) than WT cells. This deficiency was restored by transfection with gelsolin or with beta1 integrin-activating antibodies. WT cells showed robust rac activation and increased [Ca(2+)](i) during early contact with collagen beads, but Gsn(-) cells showed very limited responses. Transfected gelsolin in Gsn(-) cells restored rac activation after collagen binding. Transfection of Gsn(-) cells with active rac increased collagen binding to WT levels. Chelation of intracellular calcium inhibited collagen binding and rac activation, whereas calcium ionophore induced rac activation in WT and Gsn(-) cells. We conclude that the ability of gelsolin to remodel actin filaments is important for collagen-induced calcium entry; calcium in turn is required for rac activation, which subsequently enhances collagen binding to unoccupied alpha 2 beta 1 integrins.


Assuntos
Citoesqueleto de Actina/metabolismo , Colágeno/metabolismo , Fibroblastos/metabolismo , Gelsolina/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Cálcio/metabolismo , Células Cultivadas , Ativação Enzimática/fisiologia , Integrina alfa2beta1/metabolismo , Camundongos , Camundongos Knockout , Microscopia Confocal , Fagocitose/fisiologia , Proteínas Proto-Oncogênicas c-akt
13.
J Biol Chem ; 278(52): 53090-7, 2003 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-14561736

RESUMO

Mechanical loading of connective tissues induces the expression of extracellular matrix and cytoskeletal genes that are involved in matrix remodeling. These processes depend in part on force transmission through beta1 integrins and actin filaments, but the role of microtubules in regulating mechanotranscriptional responses is not well defined. We assessed the involvement of microtubules in the mechanotranscriptional regulation of filamin A, an actin-cross-linking protein that protects cells against force-induced apoptosis by stabilizing cell membranes. Collagen-coated magnetite beads and magnetic fields were used to apply tensile forces to cultured fibroblasts at focal adhesions. Force enhanced recruitment of alpha-tubulin and the plus end microtubule-binding protein cytoplasmic linker protein-170 (CLIP-170) at focal adhesions. Immunoprecipitation studies demonstrated no direct binding of tubulin to actin or filamin A, but CLIP-170 interacted with tubulin, filamin A, and beta-actin. The association of CLIP-170 with beta-actin was enhanced by force. Force activated the p38 mitogen-activated protein kinase, increased filamin A expression, and induced the relocation of p38 and filamin A to focal adhesions. Disruption of microtubules with nocodazole, independent of force application, enhanced filamin A expression and Sp1-mediated filamin A promoter activity, while stabilization of microtubules with Taxol inhibited force induction of both filamin A mRNA and protein. We conclude that in response to tensile forces applied through beta1 integrins and actin the microtubule network modulates mechanotranscriptional coupling of filamin A.


Assuntos
Fibroblastos/metabolismo , Microtúbulos/metabolismo , Transcrição Gênica , Actinas/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Colágeno/química , Proteínas Contráteis/metabolismo , DNA/metabolismo , Filaminas , Adesões Focais/metabolismo , Gengiva/citologia , Humanos , Immunoblotting , Magnetismo , Proteínas dos Microfilamentos/metabolismo , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas de Neoplasias , Paclitaxel/farmacologia , Reação em Cadeia da Polimerase , Testes de Precipitina , Regiões Promotoras Genéticas , Ligação Proteica , RNA/metabolismo , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fator de Transcrição Sp1/metabolismo , Estresse Mecânico , Transfecção , Tubulina (Proteína)/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
14.
Exp Cell Res ; 286(2): 366-80, 2003 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-12749864

RESUMO

Matrix remodeling by phagocytic fibroblasts is essential for growth and development but the regulatory processes are undefined. We evaluated the impact of spreading on the binding step of collagen phagocytosis with a novel culture system that more closely replicates phagocytosis in vivo than previous models. 3T3 cells were plated on collagen-coated beads, thereby loading only ventral surfaces (adhesion with spreading), or were allowed to spread on collagen films and then loaded with beads on their dorsal surfaces (adhesion without spreading). Ventral surfaces bound three-fold more beads than dorsal surfaces which was accompanied by accelerated phagosomal maturation. Arp3 and cortactin, markers of the actin-associated spreading machinery, strongly accumulated around ventrally but not dorsally loaded beads, suggesting that spreading contributes to enhanced binding of ventral surfaces. Further, ventral surfaces exhibited two-fold more free alpha2beta1 integrins, the major collagen receptors. Notably, compared to cells spread on collagen substrates, spreading cells exhibited a three-fold higher alpha2beta1 mobile fraction which was correlated with limited engagement of ventral receptors by actin filaments. Thus integrin ligation by actin filaments regulates the mobility of collagen receptors which in turn mediates the enhanced binding of collagen beads on spreading surfaces.


Assuntos
Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Colágeno/farmacologia , Técnicas de Cultura/métodos , Fibroblastos/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Células 3T3 , Citoesqueleto de Actina/metabolismo , Proteína 3 Relacionada a Actina , Animais , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/fisiologia , Adesão Celular/fisiologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Movimento Celular/fisiologia , Polaridade Celular/efeitos dos fármacos , Polaridade Celular/fisiologia , Colágeno/metabolismo , Proteínas do Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Integrina alfa2beta1/metabolismo , Camundongos , Microscopia Eletrônica de Varredura , Microesferas , Fagocitose/fisiologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia
15.
J Biol Chem ; 277(49): 47541-50, 2002 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-12324467

RESUMO

Connective tissue cells in mechanically active environments survive applied physical forces by modifying actin cytoskeletal structures that stabilize cell membranes. In fibroblasts, tensile forces induce the expression of filamin-A, a mechanoprotective actin-binding protein, but the mechanisms and protein interactions by which force activates filamin-A transcription are not defined. We found that in fibroblasts, application of tensile forces through collagen-coated magnetite beads to cell surface beta(1) integrins induced filamin-A expression. This induction required actin filaments and selective activation of the p38 mitogen-activated protein kinase. Force promoted the redistribution of p38 to the integrin/bead locus and the nucleus as well as enhanced binding of the transcription factor Sp1 to proximal, regulatory domains of the filamin-A promoter. Force application increased association of Sp1 with p38 and phosphorylation of Sp1. Transcriptional activation of filamin-A in force-treated fibroblasts was subsequently mediated by Sp1-binding sites on the filamin-A promoter. These results provide evidence for a mechanically coupled transcriptional circuit that originates at the magnetite bead/integrin locus, activates p38, tethers p38 to actin filaments, promotes binding of p38 to Sp1 in the nucleus, and induces filamin-A expression.


Assuntos
Proteínas Contráteis/metabolismo , Integrina beta1/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fator de Transcrição Sp1/metabolismo , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Proteínas Contráteis/genética , Relação Dose-Resposta a Droga , Fibroblastos/metabolismo , Filaminas , Humanos , MAP Quinase Quinase 6 , Camundongos , Proteínas dos Microfilamentos/genética , Microscopia de Fluorescência , Fosforilação , Regiões Promotoras Genéticas , Ligação Proteica , RNA/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estresse Mecânico , Fatores de Tempo , Transfecção , Raios Ultravioleta , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA