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










Base de dados
Intervalo de ano de publicação
1.
Mol Biol Cell ; 31(15): 1595-1610, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32432944

RESUMO

Tractional remodeling of collagen fibrils by fibroblasts requires long cell extensions that mediate fibril alignment. The formation of these cell extensions involves flightless I (FliI), an actin-binding protein that contains a leucine-rich-repeat (LRR), which binds R-ras and may regulate cdc42. We considered that FliI interacts with small GTPases and their regulators to mediate assembly of cell extensions. Mass spectrometry analyses of FliI immunoprecipitates showed abundant Ras GTPase-activating-like protein (IQGAP1), which in immunostained samples colocalized with FliI at cell adhesions. Knockdown of IQGAP1 reduced the numbers of cell extensions and the alignment of collagen fibrils. In experiments using dominant negative mutants, cdc42 activity was required for the formation of short extensions while R-ras was required for the formation of long extensions. Immunoprecipitation of wild-type and mutant constructs showed that IQGAP1 associated with cdc42 and R-ras; this association required the GAP-related domain (1004-1237 aa) of IQGAP1. In cells transfected with FliI mutants, the LRR of FliI, but not its gelsolin-like domains, mediated association with cdc42, R-ras, and IQGAP1. We conclude that FliI interacts with IQGAP1 and co-ordinates with cdc42 and R-ras to control the formation of cell extensions that enable collagen tractional remodeling.


Assuntos
Extensões da Superfície Celular/metabolismo , Matriz Extracelular/metabolismo , Proteínas dos Microfilamentos/metabolismo , Transativadores/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo , Proteínas ras/metabolismo , Células 3T3 , Animais , Adesão Celular , Colágeno/farmacologia , Camundongos , Modelos Biológicos , Ligação Proteica/efeitos dos fármacos , Domínios Proteicos , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas Ativadoras de ras GTPase/química
2.
Mol Biol Cell ; 29(20): 2481-2493, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30091651

RESUMO

Flightless I (FliI) is a calcium-dependent, actin severing and capping protein that localizes to cell matrix adhesions, contributes to the generation of cell extensions, and colocalizes with Ras. Currently, the mechanism by which FliI interacts with Ras to enable assembly of actin-based cell protrusions is not defined. R-Ras, but not K-ras, H-ras, or N-ras, associated with the leucine-rich region (LRR) of FliI. Mutations of the proline-rich region of R-ras (P202A, P203A) prevented this association. Knockdown of Ras GTPase-activating SH3 domain-binding protein (G3BP1) or Rasgap120 by small interfering RNA inhibited the formation of cell extensions and prevented interaction of R-ras and G3BP1 in FliI wild-type (WT) cells. Pull-down assays using G3BP1 fusion proteins showed a strong association of R-ras with the C-terminus of G3BP1 (amino acids 236-466), which also required the LRR of FliI. In cells that expressed the truncated N-terminus or C-terminus of G3BP1, the formation of cell extensions was blocked. Endogenous Rasgap120 interacted with the N-terminus of G3BP1 (amino acids 1-230). We conclude that in cells plated on collagen FliI-LRR interacts with R-ras to promote cell extension formation and that FliI is required for the interaction of Rasgap120 with G3BP1 to regulate R-ras activity and growth of cell extensions.


Assuntos
Proteínas de Transporte/metabolismo , Extensões da Superfície Celular/metabolismo , Leucina/metabolismo , Proteínas ras/metabolismo , Animais , Proteínas de Transporte/química , Bovinos , Colágeno/metabolismo , DNA Helicases/química , Camundongos , Proteínas dos Microfilamentos , Modelos Biológicos , Proteínas de Ligação a Poli-ADP-Ribose/química , Ligação Proteica , RNA Helicases/química , Proteínas com Motivo de Reconhecimento de RNA/química , Transdução de Sinais , Transativadores , Proteínas ras/química , Domínios de Homologia de src
3.
Nat Commun ; 8(1): 1530, 2017 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-29146911

RESUMO

Cytokinesis is initiated by the localized assembly of the contractile ring, a dynamic actomyosin structure that generates a membrane furrow between the segregating chromosomal masses to divide a cell into two. Here we show that the stabilization and organization of the cytokinetic furrow is specifically dependent on localized ß-actin filament assembly at the site of cytokinesis. ß-actin filaments are assembled directly at the furrow by an anillin-dependent pathway that enhances RhoA-dependent activation of the formin DIAPH3, an actin nucleator. DIAPH3 specifically generates homopolymeric filaments of ß-actin in vitro. By employing enhancers and activators, cells can achieve acute spatio-temporal control over isoform-specific actin arrays that are required for distinct cellular functions.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Citocinese/fisiologia , Proteínas dos Microfilamentos/metabolismo , Actomiosina/metabolismo , Plaquetas/fisiologia , Membrana Celular/metabolismo , Forminas , Células HeLa , Humanos , Proteínas dos Microfilamentos/genética , Microscopia de Fluorescência , Isoformas de Proteínas/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
4.
Mol Biol Cell ; 24(6): 734-47, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23325791

RESUMO

We examine how collagen substrate topography, free intracellular calcium ion concentration ([Ca(2+)]i, and the association of gelsolin with nonmuscle myosin IIA (NMMIIA) at collagen adhesions are regulated to enable collagen phagocytosis. Fibroblasts plated on planar, collagen-coated substrates show minimal increase of [Ca(2+)]i, minimal colocalization of gelsolin and NMMIIA in focal adhesions, and minimal intracellular collagen degradation. In fibroblasts plated on collagen-coated latex beads there are large increases of [Ca(2+)]i, time- and Ca(2+)-dependent enrichment of NMMIIA and gelsolin at collagen adhesions, and abundant intracellular collagen degradation. NMMIIA knockdown retards gelsolin recruitment to adhesions and blocks collagen phagocytosis. Gelsolin exhibits tight, Ca(2+)-dependent binding to full-length NMMIIA. Gelsolin domains G4-G6 selectively require Ca(2+) to interact with NMMIIA, which is restricted to residues 1339-1899 of NMMIIA. We conclude that cell adhesion to collagen presented on beads activates Ca(2+) entry and promotes the formation of phagosomes enriched with NMMIIA and gelsolin. The Ca(2+) -dependent interaction of gelsolin and NMMIIA in turn enables actin remodeling and enhances collagen degradation by phagocytosis.


Assuntos
Cálcio/metabolismo , Colágeno/metabolismo , Gelsolina/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Fagocitose/fisiologia , Actinas/metabolismo , Animais , Adesão Celular , Células Cultivadas , Colágeno/química , Fibroblastos , Adesões Focais , Humanos , Camundongos , Miosina não Muscular Tipo IIA/genética , Interferência de RNA , RNA Interferente Pequeno
5.
Am J Physiol Cell Physiol ; 295(1): C130-7, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18434624

RESUMO

Collagen phagocytosis is a crucial alpha2beta1-integrin-dependent process that mediates extracellular matrix remodeling by fibroblasts. We showed previously that after initial contact with collagen, activated Rac1 accelerates collagen phagocytosis but the Rac guanine nucleotide exchange factors (GEFs) that regulate Rac are not defined. We examined here the GEFs that regulate collagen phagocytosis in mouse fibroblasts. Collagen binding enhanced Rac1 activity (5-20 min) but not Cdc42 or RhoA activity. Analysis of collagen bead-associated proteins showed enrichment with Vav2, which correlated temporally with increased Rac1 activity. Knockdown of Vav2 prevented Rac activation, recruitment of Rac1 to collagen bead binding sites, and collagen bead binding, but knockdown of Sos-1 or beta-Pix had no effect on Rac activation or collagen binding. Vav2 was associated with the nucleotide-free Rac1 mutant (G15ARac1) after collagen binding. Collagen bead binding promoted phosphorylation of Vav2, which temporally correlated with Rac1 activation and which required Src kinase activity. Blockage of Src activity prevented collagen bead-induced Rac activation and collagen bead binding. Collectively these data indicate that Vav2 regulates the Rac1 activity associated with the binding step of collagen phagocytosis.


Assuntos
Colágeno/fisiologia , Proteínas Proto-Oncogênicas c-vav/fisiologia , Animais , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Camundongos , Células NIH 3T3 , Fagocitose , Fatores de Troca de Nucleotídeo Guanina Rho , Proteína SOS1/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Quinases da Família src/metabolismo
6.
Mol Biol Cell ; 18(8): 3026-38, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17538019

RESUMO

Phosphoinositides regulate several actin-binding proteins but their role at intercellular adhesions has not been defined. We found that phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) was generated at sites of N-cadherin-mediated intercellular adhesion and was a critical regulator of intercellular adhesion strength. Immunostaining for PI(4,5)P2 or transfection with GFP-PH-PLCdelta showed that PI(4,5)P2 was enriched at sites of N-cadherin adhesions and this enrichment required activated Rac1. Isoform-specific immunostaining for type I phosphatidylinositol 4-phosphate 5 kinase (PIP5KI) showed that PIP5KIgamma was spatially associated with N-cadherin-Fc beads. Association of PIP5KIgamma with N-cadherin adhesions was in part dependent on the activation of RhoA. Transfection with catalytically inactive PIP5KIgamma blocked the enrichment of PI(4,5)P2 around beads. Catalytically inactive PIP5KIgamma or a cell-permeant peptide that mimics and competes for the PI(4,5)P2-binding region of the actin-binding protein gelsolin inhibited incorporation of actin monomers in response to N-cadherin ligation and reduced intercellular adhesion strength by more than twofold. Gelsolin null fibroblasts transfected with a gelsolin severing mutant containing an intact PI(4,5)P2 binding region, demonstrated intercellular adhesion strength similar to wild-type transfected controls. We conclude that PIP5KIgamma-mediated generation of PI(4,5)P2 at sites of N-cadherin contacts regulates intercellular adhesion strength, an effect due in part to PI(4,5)P2-mediated regulation of gelsolin.


Assuntos
Actinas/metabolismo , Caderinas/metabolismo , Gelsolina/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Ligação Competitiva , Adesão Celular , Galinhas , Ativação Enzimática , Fibroblastos/citologia , Fibroblastos/ultraestrutura , Humanos , Isoenzimas/metabolismo , Camundongos , Células NIH 3T3 , Peptídeos/metabolismo , Ligação Proteica , Ratos , Transdução de Sinais , Proteínas rho de Ligação ao GTP/metabolismo
7.
Mol Biol Cell ; 16(11): 5175-90, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16120646

RESUMO

Collagen phagocytosis is a critical mediator of extracellular matrix remodeling. Whereas the binding step of collagen phagocytosis is facilitated by Ca2+-dependent, gelsolin-mediated severing of actin filaments, the regulation of the collagen internalization step is not defined. We determined here whether phosphatidylinositol-4,5-bisphosphate [PI(4,5)P2] regulation of gelsolin is required for collagen internalization. In gelsolin null fibroblasts transfected with gelsolin severing mutants, actin severing and collagen binding were strongly impaired but internalization and actin monomer addition at collagen bead sites were much less affected. PI(4,5)P2 accumulated around collagen during internalization and was associated with gelsolin. Cell-permeable peptides mimicking the PI(4,5)P2 binding site of gelsolin blocked actin monomer addition, the association of gelsolin with actin at phagosomes, and collagen internalization but did not affect collagen binding. Collagen beads induced recruitment of type 1 gamma phosphatidylinositol phosphate kinase (PIPK1gamma661) to internalization sites. Dominant negative constructs and RNA interference demonstrated a requirement for catalytically active PIPK1gamma661 for collagen internalization. We conclude that separate functions of gelsolin mediate sequential stages of collagen phagocytosis: Ca2+-dependent actin severing facilitates collagen binding, whereas PI(4,5)P2-dependent regulation of gelsolin promotes the actin assembly required for internalization of collagen fibrils.


Assuntos
Colágeno/metabolismo , Fibroblastos/metabolismo , Gelsolina/fisiologia , Fosfatidilinositol 4,5-Difosfato/farmacologia , Actinas/metabolismo , Animais , Cálcio/metabolismo , Dicroísmo Circular , Gelsolina/metabolismo , Camundongos , Camundongos Knockout , Mutação , Fagocitose/fisiologia , Fagossomos , Fosfatos de Fosfatidilinositol/farmacologia , Fosfotransferases (Aceptor do Grupo Álcool) , Dobramento de Proteína , Interferência de RNA , Transfecção
8.
J Biol Chem ; 276(38): 35967-77, 2001 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-11466312

RESUMO

Cells in mechanically active environments form extensive, cadherin-mediated intercellular junctions that are important in tissue remodeling and differentiation. Currently, it is unknown whether adherens junctions in connective tissue fibroblasts transmit mechanical signals and coordinate multicellular adaptations to physical forces. We hypothesized that cadherins mediate intercellular mechanotransduction by activating calcium-permeable, stretch-sensitive channels. Human gingival fibroblasts in suspension were plated on established homotypic monolayer cultures. The cells formed intercellular adherens junctions. Controlled mechanical forces were applied to intercellular junctions by electromagnets acting on cells containing internalized magnetite beads. At early but not later stages of intercellular attachment, force application visibly displaced magnetite bead-loaded cells and induced robust Ca(2+) transients (65 +/- 9.4 nm above base line). Similar Ca(2+) transients were induced by force application to anti-N-cadherin antibody-coated magnetite beads. Ca(2+) responses depended on influx of extracellular Ca(2+) through mechanosensitive channels because both Ca(2+) chelation and gadolinium chloride abolished the response and MnCl(2) quenched fura-2 fluorescence after force application. Force application induced accumulation of microinjected rhodamine-actin at intercellular contacts; actin assembly was inhibited by buffering intracellular calcium fluxes. Our results indicate that mechanical forces applied to adherens junctions activate stretch-sensitive calcium-permeable channels and increase actin polymerization. We suggest that N-cadherins in fibroblasts are intercellular mechanotransducers.


Assuntos
Caderinas/fisiologia , Sinalização do Cálcio , Células Cultivadas , Humanos , Junções Intercelulares/metabolismo , Estimulação Física
9.
J Biol Chem ; 276(34): 31969-77, 2001 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-11423540

RESUMO

Cells in mechanically active environments can activate cytoprotective mechanisms to maintain membrane integrity in the face of potentially lethal applied forces. Cytoprotection may be mediated by expression of membrane-associated cytoskeletal proteins including filamin A, an actin-binding protein that increases the rigidity of the subcortical actin cytoskeleton. In this study, we tested the hypotheses that applied forces induce the expression of filamin A specifically and that this putative protective response inhibits cell death. Magnetically generated forces were applied to protein-coated magnetite beads bound to human gingival fibroblasts, cells with constitutively low basal levels of filamin A mRNA and protein. Forces applied through collagen or fibronectin, but not bovine serum albumin or poly-l-lysine-coated beads, increased mRNA and protein content of filamin A by 3-7-fold. Forces had no effect on the expression of other filamin isotypes or other cytoskeletal proteins. This effect was dependent on the duration of force and was blocked by anti-beta(1) integrin antibodies. Force also stimulated a 60% increase in expression of luciferase under the control of a filamin A promoter in transiently transfected Rat2 fibroblasts and was dependent on Sp1 transcription factor binding sites located immediately upstream of the transcription start site. Experiments with actinomycin D-treated cells showed that the increased filamin A expression after force application was due in part to prolongation of mRNA half-life. Antisense filamin oligonucleotides blocked force-induced filamin A expression and increased cell death by >2-fold above controls. The force-induced regulation of filamin A was dependent on intact actin filaments. We conclude that cells from mechanically active environments can couple diverse signals from forces applied through beta-integrins to up-regulate the production of cytoprotective cytoskeletal proteins, typified by filamin A.


Assuntos
Proteínas Contráteis/biossíntese , Gengiva/citologia , Integrina beta1/metabolismo , Proteínas dos Microfilamentos/biossíntese , Animais , Sequência de Bases , Células Cultivadas , Proteínas Contráteis/genética , Primers do DNA , Fibroblastos/citologia , Fibroblastos/metabolismo , Filaminas , Gengiva/metabolismo , Humanos , Proteínas dos Microfilamentos/genética , Regiões Promotoras Genéticas , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição Sp1/metabolismo
10.
J Biol Chem ; 276(17): 14100-9, 2001 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-11278659

RESUMO

The immunosuppressant cyclosporin A (CsA) markedly inhibits collagen degradation by an intracellular phagocytic pathway in fibroblasts, an effect that can lead to massive gingival overgrowth. We used a collagen bead model of collagen phagocytosis to determine whether CsA inhibits internalization by blocking efflux of calcium from endoplasmic reticulum (ER) and mitochondrial calcium stores. CsA caused dose-dependent inhibition of phagocytosis of collagen-coated (but not bovine serum albumin-coated) beads. Chelation of intracellular Ca(2+) with BAPTA/AM or inhibition of Ca(2+)-ATPase of ER stores with thapsigargin reduced collagen bead phagocytosis. Measurement of intracellular calcium by ratio fluorometry showed increases in response to collagen-coated beads. Preincubation with CsA or thapsigargin caused a >3-fold decrease in intracellular calcium elevations in response to stimulation with collagen beads. Direct measurements of Ca(2+) in mitochondrial and ER stores showed that CsA only slightly inhibited collagen bead-induced discharge of calcium from mitochondria, but almost completely blocked discharge from ER stores. We reduced the numbers of mitochondria with chronic ethidium bromide treatment to test for the importance of ER/mitochondrial interactions. In these cells, CsA delayed collagen bead-induced calcium discharge from mitochondria. Collectively, these data indicate that CsA inhibits collagen phagocytosis by blocking calcium release from ER stores and may perturb functional interactions between the ER and mitochondria that regulate calcium stores.


Assuntos
Colágeno/metabolismo , Ciclosporina/farmacologia , Animais , Sítios de Ligação , Western Blotting , Cálcio/metabolismo , Células Cultivadas , Quelantes/farmacologia , Relação Dose-Resposta a Droga , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Retículo Endoplasmático/metabolismo , Inibidores Enzimáticos/farmacologia , Fibroblastos/metabolismo , Citometria de Fluxo , Humanos , Cinética , Potenciais da Membrana , Mitocôndrias/metabolismo , Fagocitose/efeitos dos fármacos , Ratos , Soroalbumina Bovina/metabolismo , Transdução de Sinais , Temperatura , Tapsigargina/farmacologia , Fatores de Tempo
11.
J Cell Sci ; 114(Pt 6): 1155-67, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11228159

RESUMO

In connective tissues, intercellular adhesion is essential for tissue morphogenesis, development and wound healing. However, the signaling mechanisms initiated by cell-cell adhesion in fibroblasts and that regulate it are not known. In this study we tested the hypothesis that intracellular calcium signaling is required to mediate intercellular adhesion between fibroblasts. Fura-2 or fluo-3 labeled human fibroblasts were used to investigate calcium homeostasis during intercellular adhesion. After contact with suspended fibroblasts there was a rise in cytosolic free calcium ([Ca2+]i) and multiple calcium oscillations in substrate-attached cells. Antibodies against the extracellular but not the cytoplasmic domain of cadherin induced a similar calcium response, indicating that these responses were initiated by cadherin binding. As shown by the near-plasma membrane Ca2+ indicator (Fura-C18) and by confocal microscopy of fluo-3-loaded cells, [Ca2+]i transients probably originated at sites of cell-cell contact. Cell-cell adhesion was dependent on both calcium influx through membrane channels and release of Ca2+ from internal calcium stores, because the calcium channel inhibitor LaCl3 or pretreatment of cells with thapsigargin significantly inhibited (>35%) cell-cell attachment. The [Ca2+]i changes induced by cell-cell adhesion were temporally correlated with increased recruitment of intercellular junctional proteins into the cytoskeleton and movement of GFP-actin to sites of cell-cell contact. [Ca2+]i responses induced by intercellular adhesion were essential for both junctional protein recruitment and the establishment of strong cell-cell contacts, as loading cells with BAPTA/AM significantly inhibited cell-cell adhesion and recruitment of cadherins and beta-catenin to the actin cytoskeleton. Actin depolymerization by cytochalasin D dramatically reduced cell-cell adhesion and recruitment of cadherins and catenin to the actin cytoskeleton. These results demonstrate that cadherin-cadherin interaction induces [Ca2+]i transients during cell-cell adhesion in fibroblasts, and these calcium signals regulate cell-cell adhesion through remodeling of cortical actin and recruitment of cadherins and beta-catenin into intercellular junctions.


Assuntos
Sinalização do Cálcio/fisiologia , Fibroblastos/fisiologia , Transativadores , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Caderinas/metabolismo , Cálcio/metabolismo , Adesão Celular/fisiologia , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Citoesqueleto/fisiologia , Fibroblastos/citologia , Humanos , Líquido Intracelular/metabolismo , Peptídeos/metabolismo , beta Catenina
12.
J Cell Sci ; 114(Pt 1): 119-129, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11112696

RESUMO

In physiological conditions, collagen degradation by fibroblasts occurs primarily via phagocytosis, an intracellular pathway that is thought to require collagen receptors and actin assembly for fibril internalization and degradation. Currently it is unclear which specific steps of collagen phagocytosis in fibroblasts involve actin filament assembly. As studies of phagocytosis in fibroblasts are complicated by the relatively slow rate of particle internalization compared to professional phagocytes, we have examined the role of collagen receptors and actin only in the initial collagen binding step. Prior to the binding of collagen-coated fluorescent beads by human gingival fibroblasts, a cell type that is avidly phagocytic in vitro, cells were treated with cytochalasin D (actin filament barbed-end capping) or swinholide A (actin dimer sequestering and severing) or latrunculin B (actin monomer sequestering). Bead binding and immunostaining of (alpha)(2)(beta)(1) and (alpha)(3)(beta)(1) integrin collagen receptors were measured by flow cytometry. After 1-3 hours of coincubation with beads, cytochalasin D or swinholide A eliminated actin filaments stained by rhodamine-phalloidin and inhibited collagen bead binding (reductions of 25% and 50%, respectively), possibly because of cell rounding and restricted interactions with beads. In contrast, latrunculin enhanced binding dose-dependently over controls (twofold at 1 microM) and induced the formation of brightly staining aggregates of actin and the retention of long cytoplasmic extensions. Latrunculin also reduced surface (beta)(1), (alpha)(2) and (alpha)(3) integrin staining up to 40% in bead-free and bead-loaded cells, indicating that latrunculin enhanced collagen receptor internalization. As determined by fluorescence recovery after photobleaching, latrunculin increased the mobility of surface-bound (beta)(1) integrin. The stimulatory effect of latrunculin on collagen bead binding was reduced to control levels by treatment with a (beta)(1) integrin inactivating antibody while a (beta)(1) integrin blocking antibody abrogated both bead binding and the latrunculin-induced stimulation. Immunoblotting of bead-associated proteins showed that latrunculin completely eliminated binding of (beta)-actin to collagen beads but did not affect (beta)(1) integrin binding. These data indicate that latrunculin-induced sequestration of actin monomers facilitates the disengagement of actin from (beta)(1) integrin receptors, increases collagen bead binding and enhances collagen receptor mobility. We suggest that these alterations increase the probability of adhesive bead-to-cell interactions.


Assuntos
Actinas/metabolismo , Colágeno/metabolismo , Fibroblastos/fisiologia , Integrinas/metabolismo , Fagocitose/fisiologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Humanos , Receptores de Colágeno , Tiazóis/farmacologia , Tiazolidinas
13.
J Biol Chem ; 275(45): 35432-41, 2000 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-10945978

RESUMO

Intracellular collagen degradation by fibroblasts is an important but poorly understood pathway for the physiological remodeling of mature connective tissues. The objective of this study was to determine whether gingival fibroblasts that express endogenous alpha(2)beta(1) integrin, the collagen receptor, would exhibit the cellular machinery required for phagosomal maturation and collagen degradation. There was a time-dependent increase of collagen bead internalization and a time-dependent decrease of bead-associated alpha(2)beta(1) integrin after initial bead binding. beta-Actin and gelsolin associated transiently with beads (0-30 min) followed by LAMP-2 (60-240 min) and cathepsin B (30-240 min). Cytochalasin D prevented phagosome formation and also prevented the sequential fusion of early endosomes with lysosomes. Collagen bead-associated pH was progressively reduced from 7.25 to 5.4, which was contemporaneous with progressive increases in degradation of bead-associated collagen (30-120 min). Concanamycin blocked acidification of phagolysosomes and collagen degradation but not phagosome maturation. Phagosomal acidification was partly dependent on elevated intracellular calcium. These studies demonstrate that the cellular machinery required for intracellular collagen degradation in fibroblasts closely resembles the vacuolar system in macrophages.


Assuntos
Colágeno/metabolismo , Fibroblastos/metabolismo , Macrolídeos , Fagossomos/fisiologia , Actinas/metabolismo , Adolescente , Animais , Antibacterianos/farmacologia , Antígenos CD/metabolismo , Cálcio/metabolismo , Cálcio/farmacologia , Catepsina B/metabolismo , Criança , Citocalasina D/farmacologia , Endossomos/metabolismo , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Fluoresceína-5-Isotiocianato/farmacologia , Gelsolina/metabolismo , Gengiva/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Integrinas/metabolismo , Proteínas de Membrana Lisossomal , Lisossomos/metabolismo , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Microscopia Eletrônica , Microscopia de Fluorescência , Inibidores da Síntese de Ácido Nucleico/farmacologia , Receptores de Colágeno , Suínos , Fatores de Tempo
14.
Am J Pathol ; 155(6): 2087-99, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10595938

RESUMO

Myofibroblasts are important but transient mediators of normal wound contraction and are characterized phenotypically by their high levels of alpha-smooth-muscle actin (SMA). During wound maturation, these cells disappear. We have examined the mechanisms that lead to myofibroblast deletion in a fibroblast culture model. Transforming growth factor-beta (TGF-beta) was used to increase SMA content in gingival fibroblasts (three- to sixfold). After replating TGF-beta-induced cells at low density with serum, there was a fivefold decrease in SMA protein content, SMA protein synthesis, and SMA mRNA as cells proliferated. These reductions were due to reduced SMA mRNA stability. For TGF-beta-induced cells plated at high density without serum (ie, quiescent conditions), protein content was reduced by only 20% over 12 days. TGF-beta protected SMA-positive cells against apoptosis in serum-free cultures. Those cells that were protected against apoptosis exhibited well-developed stress fibers enriched in SMA. We conclude that, in quiescent myofibroblasts, SMA protein turnover is slow, and cells are long-lived. In proliferative conditions SMA protein and mRNA turn over quickly, and the myofibroblast phenotype dissipates. The reduced apoptosis of myofibroblasts in quiescent conditions is due in part to the organization of SMA into stress fibers.


Assuntos
Actinas/biossíntese , Apoptose , Fibroblastos/citologia , Fibroblastos/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Northern Blotting , Western Blotting , Divisão Celular , Células Cultivadas , Meios de Cultura , Citometria de Fluxo , Gengiva/citologia , Humanos , Microscopia de Fluorescência , Músculo Liso , Fenótipo , Testes de Precipitina , RNA Mensageiro/análise
15.
Exp Cell Res ; 250(1): 155-67, 1999 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-10388529

RESUMO

Gelsolin is an abundant actin binding protein that mediates the rapid remodeling of cortical actin filaments through severing, capping, and nucleating activities. Most of the attention on the intracellular function of gelsolin has focused on the remodeling of the cortical actin meshwork but the localization of gelsolin to other regions of the cell suggests that it may have other important functions elsewhere. In cultured fibroblasts, gelsolin is heavily enriched in stress fibers, where its function in these contractile organelles is unknown. To study gelsolin function during stress fiber formation and cell contraction, we first assessed gelsolin levels in stress fiber preparations from lysophosphatidic acid (LPA)-treated human fibroblasts. LPA induced a large, time-dependent, calcium-independent increase of actin, gelsolin, alpha-actinin, and tropomyosin in stress fiber preparations. A microinjected gelsolin antibody that inhibits severing by gelsolin reduced stress fibers. Anti-sense-transfected gelsolin-depleted 3T3 cell lines treated with LPA after serum starvation required approximately 6 h to form stress fibers and focal adhesions, in contrast to control lines transfected with vector only, which formed stress fibers 15 min after addition of LPA. In cells microinjected with the gelsolin antibody that inhibits severing, Mg-ATP-induced cell contraction was greatly reduced in approximately 90% of injected cells compared to cells injected with an irrelevant antibody. Gelsolin-depleted cells were incapable of collagen gel contraction and showed no apparent Mg-ATP-induced cell contraction compared to cell lines transfected with vector only. The involvement of gelsolin in cell contraction and remodeling of collagen gels suggests a novel role for gelsolin in stress fiber-dependent cell function.


Assuntos
Citoesqueleto de Actina/fisiologia , Gelsolina/fisiologia , Células 3T3 , Citoesqueleto de Actina/metabolismo , Animais , Gelsolina/metabolismo , Humanos , Lisofosfolipídeos/farmacologia , Camundongos
17.
Am J Pathol ; 154(3): 871-82, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10079265

RESUMO

Wound contraction is mediated by myofibroblasts, specialized fibroblasts that appear in large numbers as the wound matures and when resistance to contractile forces increases. We considered that the regulation of myofibroblast differentiation by wound-healing cytokines may be dependent on the resistance of the connective tissue matrix to deformation. We examined transforming growth factor-beta1 (TGF-beta1) induction of the putative fibroblast contractile marker, alpha-smooth muscle actin (alpha-SMA), and the regulation of this process by the compliance of collagen substrates. Cells were cultured in three different types of collagen gels with wide variations of mechanical compliance as assessed by deformation testing. The resistance to collagen gel deformation determined the levels of intracellular tension as shown by staining for actin stress fibers. For cells plated on thin films of collagen-coated plastic (ie, minimal compliance and maximal intracellular tension), TGF-beta1 (10 ng/ml; 6 days) increased alpha-SMA protein content by ninefold as detected by Western blots but did not affect beta-actin content. Western blots of cells in anchored collagen gels (moderate compliance and tension) also showed a TGF-beta1-induced increase of alpha-SMA content, but the effect was greatly reduced compared with collagen-coated plastic (<3-fold increase). In floating collagen gels (high compliance and low tension), there were only minimal differences of alpha-SMA protein. Northern analyses for alpha-SMA and beta-actin indicated that TGF-beta1 selectively increased mRNA for alpha-SMA similar to the reported protein levels. In pulse-chase experiments, [35S]methionine-labeled intracellular alpha-SMA decayed most rapidly in floating gels, less rapidly in anchored gels, and not at all in collagen plates after TGF-beta1 treatment. TGF-beta1 increased alpha2 and beta1 integrin content by 50% in cells on collagen plates, but the increase was less marked on anchored gels and was undetectable in floating gels. When intracellular tension on collagen substrates was reduced by preincubating cells with blocking antibodies to the alpha2 and beta1 integrin subunits, TGF-beta1 failed to increase alpha-SMA protein content in all three types of collagen matrices. These data indicate that TGF-beta1-induced increases of alpha-SMA content are dependent on the resistance of the substrate to deformation and that the generation of intracellular tension is a central determinant of contractile cytoskeletal gene expression.


Assuntos
Actinas/metabolismo , Colágeno/fisiologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Músculo Liso/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Actinas/antagonistas & inibidores , Actinas/genética , Adolescente , Anticorpos/farmacologia , Células Cultivadas , Criança , Complacência (Medida de Distensibilidade) , Fibroblastos/fisiologia , Géis , Humanos , Integrinas/metabolismo , RNA Mensageiro/metabolismo , Fator de Crescimento Transformador beta/imunologia
18.
J Biol Chem ; 271(34): 20516-23, 1996 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-8702793

RESUMO

Gelsolin nucleates actin filament assembly, blocks the fast-exchanging ends of actin filaments, and severs filaments, processes that may play an important role in cell motility. We studied the relationship between cell migration, gelsolin content, and actin severing activity in human gingival fibroblasts. These cells were keratin negative and desmin negative but expressed vimentin and myosin II. Cells were separated by their ability to migrate in response to a chemoattractant stimulus. Northern analysis of mRNA, [35S]methionine incorporation into immunoprecipitated gelsolin, immunoblots of cell lysates, and quantitative confocal microscopy showed 1.4-2-fold higher levels of gelsolin in nonmigrant compared with migrant cells. Because the concentration of intracellular gelsolin did not appear to be a central determinant of cell migration, we assessed its requirement for motility. Cells that were electroinjected with a gelsolin antibody that inhibits actin severing by gelsolin in vitro showed a 72% reduction of the number of migrant cells compared with controls treated with an irrelevant antibody. Cells that were electroinjected with free gelsolin exhibited a 33% increase in migration compared with cells electroinjected with bovine serum albumin. Compared with nonmigrant cells, migrant cells contained abundant free gelsolin and exhibited gelsolin-dependent F-actin severing activity, which required Ca2+. Serum stimulation of cell migration required increases in [Ca2+]i because incubation with 3 microM 1,2-bis-(o-aminophenoxy)-ethane-N,N, N',N'-tetraacetic acid tetra(acetoxymethyl)-ester blocked calcium flux and cell migration. Serum also stimulated the recruitment of gelsolin into the supernatants of lysates from migrant but not from nonmigrant cells. In fibroblasts, gelsolin concentration alone does not apparently determine migratory capacity. Instead, the Ca2+-dependent actin severing activity of free gelsolin appears to be a major determinant of cell migration.


Assuntos
Citoesqueleto de Actina/ultraestrutura , Actinas/fisiologia , Movimento Celular , Fibroblastos/citologia , Gelsolina/metabolismo , Citoesqueleto de Actina/fisiologia , Cálcio/metabolismo , Compartimento Celular , Quimiotaxia , Desmina/metabolismo , Gengiva/citologia , Humanos , Queratinas/metabolismo , Microscopia Confocal , Fosfatidilinositóis/fisiologia
19.
J Biol Chem ; 270(11): 6042-9, 1995 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-7890736

RESUMO

Interleukin-1 (IL-1) is an important mediator of inflammation and also modulates fibroblast metabolism. To assess mechanisms of IL-1-induced signal transduction and calcium flux, early passage human fibroblasts were loaded with fura2/AM. Cells grown on coverslips exhibited dose-dependent [Ca2+]i responses that were maximal at 10(-8) M IL-1 beta with time to maximum flux of 50 s. Cells incubated with anti-Type 1-IL-1 receptor antibody exhibited a 45 nM increase in [Ca2+]i above baseline but demonstrated no calcium response after IL-1 beta treatment. Incubation with EGTA (5 mM) or thapsigargin (1 microM) caused 75% and 37% reductions, respectively, in the IL-1-induced [Ca2+]i increase, suggesting that extracellular Ca2+ predominates in IL-1-stimulated calcium flux. Cells in suspension did not exhibit [Ca2+]i responses to IL-1 beta. The relationship between [Ca2+]i signaling and focal adhesions was examined by plating cells on fibronectin or poly-L-lysine, conditions that either permitted or blocked the formation of focal adhesions. Cells on fibronectin exhibited co-distribution of immunostaining for talin, vinculin, IL-1 receptor, and focal adhesion kinase (pp125fak) in focal adhesions and demonstrated [Ca2+]i responses with 10(-8) M IL-1 beta. Cells on poly-L-lysine or cells in suspension did not exhibit co-distribution of pp125fak, IL-1 receptor, and focal adhesion proteins and did not exhibit calcium flux. The dependence of IL-1-stimulated [Ca2+]i responses on tyrosine kinases was examined first by treating cells with genistein, a selective inhibitor of tyrosine kinases. Genistein (100 microM) completely blocked [Ca2+]i responses to 10(-8) M IL-1, whereas its inactive analogue genistin was not inhibitory. Second, fibroblasts lysates were immunoprecipitated with an antiphosphotyrosine antibody and the lysates were Western-blotted with an anti-pp125fak antibody. Cells grown on fibronectin and stimulated with IL-1 exhibited tyrosine phosphorylation of pp125fak whereas untreated cells or cells grown on poly-L-lysine and treated with IL-1 showed no reaction. Fibroblasts electroinjected with anti-pp125fak monoclonal antibody showed no [Ca2+], response, whereas cells treated with an irrelevant antibody exhibited a normal [Ca2+]i response. Collectively, these data indicate that fibroblasts require substrate attachment and clustering of IL-1 receptors to focal adhesions for IL-1-induced [Ca2+]i responses. Calcium fluxes are mediated through tyrosine kinases whose substrates include pp125fak. These studies therefore demonstrate that activation of intracellular signaling pathways by IL-1 is dependent on IL-1 receptor-cytoskeletal protein interactions.


Assuntos
Cálcio/metabolismo , Adesão Celular/efeitos dos fármacos , Gengiva/metabolismo , Interleucina-1/farmacologia , Receptores de Interleucina-1/fisiologia , Moléculas de Adesão Celular/análise , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Eletroporação , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Citometria de Fluxo , Imunofluorescência , Corantes Fluorescentes , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Fura-2/análogos & derivados , Humanos , Cinética , Microscopia Confocal , Fosforilação , Proteínas Tirosina Quinases/análise , Proteínas Tirosina Quinases/metabolismo , Receptores de Interleucina-1/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Espectrometria de Fluorescência , Talina/análise , Fatores de Tempo , Vinculina/análise
20.
J Cell Physiol ; 161(2): 187-200, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7962103

RESUMO

Calcium transients in single, human gingival fibroblasts were studied after mechanical stretching of flexible culture substrates. A model system was developed to reproducibly stretch and rapidly (<1 sec) refocus cells in the same focal plane so that changes in the concentration of free intracellular calcium ions ([Ca2+]i) were monitored without delay. Attached cells were grown on flexible bottom Petriperm dishes, loaded with fura-2/AM, and stretched by 1% or 2.8% of substrate area. The stretch caused no significant cell detachment or membrane lesions. A 1% stretch induce no calcium response, but a 2.8% stretch stimulated an initial calcium transient and the subsequent generation of [Ca2+]i oscillations of up to 2,000 sec. At 1% stretch, there was no calcium response. Cell shape and plating time were important determinants in the calcium response to mechanical stimulation: the responder cells were small and round without long processes. Major calcium transients were inhibited completely by 5 mM EGTA or by 10 microM gadolinium ions, by 50 microM nifedipine, or 250 microM verapamil, suggesting an influx of calcium through stretch-activated (SA) channels and L-type calcium channels. Depolarization by high KCl (144 mM) in the extracellular medium enhanced the amplitude of calcium transients by 54%. Calcium oscillations were not inhibited by preincubation with thapsigargin, caffeine, cholera toxin, staurosporine or 1-(5-isoquinolinesulfonyl)-2-methylpiperazine (H-7), indicating that IP3 sensitive pools, IP3 insensitive pools, GS alpha subunits, and protein kinase C, respectively, were not involved in the generation of calcium oscillations. Pretreatment with genistein, a specific tyrosine kinase inhibitor or cytochalasin D, an inhibitor of actin polymerization, or pertussis toxin, an inhibitor of Gi alpha and G(o) alpha subunits, completely abolished calcium transients and oscillations. These results indicate that Ca2+ flux due to mechanical stretching is likely mediated through SA ion channels and is dependent on tyrosine kinases, pertussis toxin-sensitive subunits of G-proteins, and actin filaments.


Assuntos
Cálcio/metabolismo , Fibroblastos/metabolismo , Membranas Intracelulares/metabolismo , Actinas/fisiologia , Canais de Cálcio/metabolismo , Polaridade Celular , Células Cultivadas , Espaço Extracelular/metabolismo , Proteínas de Ligação ao GTP/fisiologia , Humanos , Oscilometria , Concentração Osmolar , Estimulação Física , Proteína Quinase C/fisiologia , Proteínas Tirosina Quinases/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...