Keratinocyte migration is an absolute requirement for correct epithelialization during the process of wound healing. This process requires changes in extracellular matrix ligand expression as well as changes in ligand-binding affinity of the corresponding cellular integrins. In this study, we attempt to understand the role of laminin 5 in migration by investigating the integrin-mediated interactions of migrating keratinocytes with their newly synthesized laminin 5. We chose to induce migration of freshly isolated NHK in vitro by exposing them to TGF-β1 which, in addition to promoting epithelial cell migration, is also known to prevent cell proliferation. This important feature allowed the study to be focused on cell migration without interfering with cell proliferation. We confirm that keratinocyte migration on plastic, fibronectin or collagen IV substrates requires endogenous laminin 5 deposition, which is predominantly detected under its unprocessed form. Despite a crucial role for laminin 5 in migration, we show that this process is accompanied by a significant decrease in adhesion to purified laminin 5. Moreover, we provide evidence that the α3β1 integrin interaction with newly synthesized laminin 5 renders the cells more adherent and retards migration. Conversely, we provide evidence that the α2β1 integrin-laminin 5 interaction is absolutely required for keratinocyte migration and that the α2β1 integrin is responsible for cell spreading on laminin 5. Finally, we demonstrate that the α2β1 integrin binding to laminin 5 occurs within the short arm of the γ2 subunit.

Interactions between cells and the extracellular matrix (ECM) are critical for many biological events and involve cell membrane receptors of the integrin family. Integrins are transmembrane receptors implicated in mediating cell-substratum attachment or cell-cell adhesion, and in transducing signals that regulate such diverse processes as growth, differentiation and migration.

In the epidermis of the skin, basal keratinocytes adhere to the basement membrane through integrins. Human keratinocytes express a number of integrins, including α2β, α3β and α6β restricted to the basal and suprabasal, proliferative cell layer, both in the epidermis and in stratified culture of keratinocytes (Carter et al., 1990a; Hertle et al., 1991; Marchisio, 1991). In the epidermis, α6β integrin is a component of hemidesmosomes (HD) (for a review, see Nievers et al., 1999). It mediates keratinocyte adhesion to the basement membrane by binding to laminin 5; this interaction is necessary for HD stability (Langhofer et al., 1993; Ryan et al., 1999). The α2β and α6β integrins are localized at the lateral plasma membranes and have been proposed to cooperate in cell-cell interactions (Kaufmann et al., 1989; Carter et al., 1990b; Symington et al., 1993).

According to tissue needs, the strength of attachment is subject to up- or downregulation during, for example, commitment to terminal differentiation or epidermal regeneration. In these situations basal cells detach from the basement membrane and migrate either into the suprabasal layers (Adams and Watt, 1990) or laterally (Larjava et al., 1993). These processes appear to involve changes both in the ligand-binding activities of certain integrins and in the expression of integrins on the cell surface. During wound healing, migration starts within hours after injury and continues until the epithelial surface is intact so as to finally regenerate the basement membrane (reviewed in Clark, 1996). Keratinocytes from the edges of the wound and within skin appendage structures migrate over a provisional matrix rich in newly synthesized fibronectin and laminin 5 (Larjava et al., 1993). Integrin polarization is lost and keratinocytes enlarge their integrin repertoire and modify their distribution. The appearance of the α6β integrin and a strong increase in the αvβ5 integrin are the major events occurring in the migratory region of the wound (Cavani et al., 1993; Juhasz et al., 1993; Larjava et al., 1993). Integrins α6β4 and α2β move from a lateral to a basal location in contact with ECM. The α6β4 integrin becomes more evenly distributed and appears along the entire cell surface of migrating keratinocytes in which HD are absent (Kurpakus et al., 1991).

Although the role of fibronectin in epidermal wound healing has been clearly demonstrated (for a review, see Humphries, 1991), it is certainly not the only ECM protein involved in this process. Dermal collagen I has been proposed to play a crucial role in keratinocyte migration by mediating induction of collagenase-1 through interaction with α2β integrins (Pilcher et al., 1997). The resulting cleavage of collagen I by collagenase-1 was proposed to be needed to initiate cell movement (Pilcher et al., 1997). Among basement membrane components involved in cellular interactions, laminin 5 is likely to play a role during cell migration. Laminin 5 is the major component of anchoring filaments of the skin and is a composed of the heterotrimeric association of the α3, β3 and γ2 laminin chains (Rousselle et al., 1991). It supports cell adhesion via interaction of its α? C-terminal extremity, called G-domain, with both the α6β4 and α6β4 integrins (Carter et al., 1991; Sonnenberg et al., 1993; Rousselle and Aumailley, 1994; Mizushima et al., 1997). Laminin 5 is synthesized as a 460 kDa precursor that undergoes specific processing to smaller forms of 440 and 400 kDa rapidly after secretion (Marinkovich et al., 1992). The size reduction is a result of a carboxy-terminal truncation of the 200 kDa α3 chain into a 165 kDa polypeptide (Goldfinger et al., 1998), and by shortening the 155 kDa γ2 chain at the amino terminus into a 105 kDa polypeptide (Vailly et al., 1994). A larger truncation has been reported for the γ2 chain synthesized by a rat tumour cell line (Giannelli et al., 1997).

Immunohistochemical and in situ hybridization studies have shown induction of laminin 5 expression in migrating keratinocytes during wound healing (Kurpakus et al., 1991; Larjava et al., 1993; Ryan et al., 1994; Lampe et al., 1998; Salo et al., 1999) or in malignant cells located at the invasion front of human carcinomas (Pyke et al., 1994). Several lines of evidence indicate that efficient migration of normal human keratinocytes (NHK) is dependent on endogenously deposited laminin 5 (Zhang and Kramer, 1996; Qin and Kurpakus, 1998; Goldfinger et al., 1999; Nguyen et al., 2000). Moreover, studies indicate that laminin 5 containing the 200 kDa α? chain is capable of inducing cell migration while impeding hemidesmosome assembly (Goldfinger et al., 1998; Goldfinger et al., 1999). Other studies provided evidence that processed-α3 containing-laminin 5 function in the nucleation of hemidesmosome assembly and as an adhesive factor that retards cell motility (Baker et al., 1996; Langhofer et al., 1993; O’Toole et al., 1997; Goldfinger et al., 1998).

This regulation, as well as the up- and downregulation of the strength of attachment occurring during cell migration, is likely to be modulated by external mediators such as growth factors and cytokines. A cytokine with a broad spectrum of effects in cell-matrix interactions is the transforming growth factor-β (TGF-β) peptide family (for a review, see Massague, 1990). In addition to controlling cell growth, these molecules play an important role in the remodelling of tissue by regulating the transcription of a wide spectrum of ECM proteins, increasing their production while decreasing their proteolysis and modulating their interactions with cellular integrin receptors. Moreover, TGF-β promotes the migration of the majority of cell types that participate in repair processes. Indeed, TGF-β1 has the ability to induce keratinocyte migration in vitro by regulating integrin expression (Zambruno et al., 1995). Moreover, it was shown to induce upregulation of the laminin α3 mRNA (Korang et al., 1995; Virolle et al., 1998).

In this study, we attempt to understand the function of laminin 5 during NHK migration. Since NHK have no tendency to migrate during normal culture conditions, we have constrained them to migrate by exposing them to TGF-β1. In order to exclude any artifact caused by the use of TGF-β1, we have verified our findings with untreated cells. We show that migration is accompanied by a significant decrease in integrin-mediated adhesion to laminin 5. Moreover, we provide evidence that the α6β4 integrin does not promote migration while the α2β integrin is absolutely necessary. Finally, we demonstrate that the α2β integrin binds to the short arm of the laminin 5 γ2 subunit.

Cell culture

NHK cultures were established from foreskin as previously described (Rousselle et al., 1991) and subcultured in Keratinocyte Serum Free Medium K-SFM (Gibco BRL, Cergy Pontoise, France). Cells were harvested for subculturing or for subsequent experiments using 0.05% trypsin and 0.02% EDTA in phosphate-buffered saline, pH 7.4 (PBS). NHK were used between passages 1-3. Cell culture reagents were from Seromed-Biochrom (Polylabo, Strasbourg, France) and plasticware was from Falcon (Dutscher, Brumath, France). NHK were harvested at a cell density of 1×105 cells/cm2, and after reaching subconfluency they were incubated in SFM for 24 hours in the presence or absence of 10 ng/ml of human platelet-derived purified TGF-β1 (R&D Systems, Abingdon, UK).

Preparation of laminin 5 matrices

After the cells were grown in the presence or absence of TGF-β1, the medium was removed and the cells were washed in sterile PBS. The cells were removed by treating them for 5 minutes in sterile 20 mM NH4OH (Gospodarowicz, 1984). The matrix was washed three times in sterile water and was then removed from the substratum by solubilization in 0.325 M Tris-HCl, pH 6.9, 25% glycerol, 10% SDS with or without 5% β-mercaptoethanol, for electrophoretic analysis.

Cell adhesion substrates and anti-integrin antibodies

Human plasma fibronectin and mouse EHS collagen IV were purchased from Becton Dickinson (Le Pont de Claix, France). EHS laminin 1 was graciously provided by Prof. Aumailley and human laminin 5 was purified from the culture medium of human SCC25 cells as previously described (Rousselle and Aumailley, 1994). Characterization of laminin 5 monoclonal antibody (mAb) BM165 (anti-α3chain) and polyclonal antibody (pAb) 4101 was described elsewhere (Rousselle et al., 1991; Marinkovich et al., 1992) and pAb L132 was raised against purified native laminin 5. mAb D4B5 against domain III of the γ2 chain was purchased from Chemicon (Euromedex, Souffelweyersheim, France). pAbs to thrombospondin (Ab-8) and collagen XVII (L 766P) were provided by Dr Clezardin (Inserm U403, Lyon, France) and Dr Meneguzzi (Inserm U385, Nice, France), respectively. mAb (T2H5) to tenascin was purchased from Chemicon and mAb NP32 to the NC1 domain of type VII collagen was provided by Prof. Burgeson (CBRC, Charlestown, USA). The following function-blocking mAbs against integrin subunits were used: P4C10 against β1, P1E6 against α2, P1B5 against α3, P1D6 against α5, all from Telios (Gibco BRL); CS-1 against α3 from Chemicon and GoH3 against α6 (Genosys, Becton Dickinson). Nonfunction-blocking mAb A3×8 to the α3 integrin subunit was graciously provided by Dr Hemler (Dana Farber Cancer Institute, Boston, USA).

Cell adhesion and inhibition assays

Multiwell tissue culture plates (96-wells, Costar, Dutscher) were coated with the indicated concentrations of laminin 5, fibronectin and collagen IV substrates by overnight adsorption at 4°C. After saturation of the wells with 1% BSA (fraction V, Sigma Chimie, France), the plates were immediately used for cell adhesion assays (3×?? 5 cells/well) in serum-free medium as detailed previously (Rousselle and Aumailley, 1994). The extent of adhesion was determined after fixation of the adherent cells with 1% glutaraldehyde in PBS, by staining with 0.1% Crystal Violet, and color reading at 570 nm with an ELISA reader (MR5000 Dynatech, Guernsey, Channel Islands). A blank value corresponding to BSA-coated wells was automatically subtracted. For all experiments, each assay point was determined in triplicate.

Cell locomotion assay

The lower face of the nucleopore filters (pore size: 8 μm) present in Transwell chambers (6 wells; Costar, through Dutscher) was coated or not with fibronectin, collagen IV or laminin 5 (10 μg/ml) and post-coated with BSA. Cells in monolayers were radioactively labeled by incubation with 35S-methionine (NEN, Les Ulis, France) for 1 hour in methionine-free SFM. Labelled cells were seeded in the upper part of the chambers (3×105 cells/well) in SFM with 0.2% BSA and allowed to migrate for 6 hours. For cell migration perturbation experiments with cycloheximide (Sigma) or various antibodies, suspended cells were mixed with cycloheximide (25 μg/ml) for 30 minutes at room temperature or dilutions of antibody prior to being seeded in the transwell. At the end of the assay, cells on the upper face of the nucleopore filters were manually removed except for a set of filters used for the determination of total cell number. Radioactivity associated with the filters was measured in a scintillation counter. The number of cells that had migrated was expressed as a percentage of radioactivity associated with the lower face versus both faces of the filters. Radioactivity associated with BSA-coated filters was subtracted from the values.

Immunofluorescence staining

Sterile round glass coverslips deposited on the bottom of 24-well plates (Costar, Dutscher) were seeded with 1×105 keratinocytes. After the cells reached subconfluency, they were incubated in SFM for 24 hours in the presence or absence of 10 ng/ml of TGF-β1. After washing the wells with PBS, the cells were fixed with 2% paraformaldehyde in PBS for 15 minutes, permeabilized with 0.2% Triton X-100 for 1 minute, rinsed several times with PBS, and incubated for 45 minutes with the first antibody. Cy3-conjugated second antibodies against mouse or rabbit immunoglobulins (Jackson, distributed through Immunotech) were applied for another 45 minutes. In the case of double immunolabelling, the procedure was followed with an additional 45 minutes incubation with pAb L132, followed by the appropriate FITC-conjugated second antibody (Biosys, Compiègne, France). Glass coverslips were mounted onto slides in FluoPrep mounting medium (BioMérieux, Marcy l’Etoile, France) and cells were observed by epifluorescence with an Axiophot Zeiss microscope.

In vitro binding assay on immobilized laminin 5

Cell lysates containing α2β integrin were prepared from detergent extracts of either cultured human keratinocytes or fibroblasts. The cells were washed in PBS and solubilized in 50 mM Tris-HCl buffer, pH 7.4, containing 50 mM n-octyl-1-thio-β-D-glucopyranoside (Roche Diagnostics, Meylan, France) and 2 mM phenylmethylsulfonyl fluoride (PMSF). After 10 minutes, the extract was centrifuged at 14,000 g for 15 minutes and the supernantant was dialysed against 50 mM Tris-HCl buffer, pH 7.4, 0.2 M NaCl (TBS) containing 2 mM MgCl2 and 1 mM MnCl2. α2β integrin binding assays on immobilized laminins were performed as followed. Multiwell plates (96 wells, Greiner, Dutcher) were incubated with a concentration of laminin 1 or 5 varying from 20 to 100 μg/ml dissolved in 20 mM Na2Co3, pH 9.2, for 18 hours at 4°C. The wells were then blocked with 5 mg/ml BSA in TBS for 1 hour at room temperature. BSA was removed and the soluble cell lysate added for 2 hours at room temperature. After the wells were washed three times with TBS 0.02% Tween 20 containing 2 mM MgCl2, 1 mM MnCl2, they were exposed to a constant amount of diluted pAb against human α2β integrin (Chemicon International, Euromedex, Mundolsheim, France) followed by a typical enzyme-immunoassay reaction with peroxidase-conjugate of donkey specific antirabbit immunoglobulin (Jackson Immuno Research Laboratories, through Interchim, Montlucon, France) as second antibody and 2,2′-azino-bis(3-ethylbenthiazoline-6-sulfonic acid) as the chromogenic substrate. Color yields were determined at 405 nm in an ELISA reader. Negative control values, from wells in which the soluble ligands were omitted, were subtracted from the binding data. For inhibition assays, soluble cell lysates and immobilized proteins were incubated with inhibitors for 1 hour at room temperature before being exposed.

Skin wound specimens

The 5-day-old-wounding skin biopsy was provided by the Centre des Grands Brûlés, Pavillon I-5, Hôpital Edouard Hérriot, Lyon (France). It was sampled from the back of a 2nd degree burn patient subject to spontaneous wounding. The 5 mm2 biopsy was taken during a dressing change. After a rapid wash in PBS, it was immediately frozen in Tissue-Tek OCT Compound (Sakura Finetek, Poly Labo) and stored at –70°C. Consecutive frozen sections of 8 μm thickness were prepared using a Reichert-Jung Leica cryostat at –20°C and placed on glass slides. Sections were then processed for indirect immunofluorescence as described above and finally incubated with Trypan Blue prior mounting with FluoPrep mounting medium.

SDS-PAGE, western immunoblotting

Protein preparations were separated by SDS-PAGE using 5% or 3%-5% acrylamide gels. Proteins were either stained with Coomassie Brilliant Blue or transferred to nitrocellulose filters for western blotting according to standard procedures. Molecular mass markers were from Bio-Rad Laboratories (Ivry, France).

The TGF-β1-induced migratory phenotype of normal keratinocytes is accompanied by a decreased adhesion to purified laminin 5

In agreement with previous reports (Boland et al., 1996; Zicha et al., 1999), we have found that TGF-β1 increased the cell motility in vitro. Cell migration assays, using Transwell chambers, demonstrated that NHK ability to migrate was greatly enhanced. We tested the ability of cells to migrate towards substrates such as laminin 5, fibronectin and collagen IV).Fibronectin was the best migrating substrate for NHK whilst there was no migration toward purified laminin 5 (Fig. 1). Although TGF-β1 treatment did not enhance cell migration towards purified laminin 5, it considerably increased cell migration towards fibronectin and collagen IV.

Fig. 1.

Cell migration assay. Keratinocyte migration toward purified laminin 5 (LN5, 10 μg/ml), fibronectin (FN, 10 μg/ml) and collagen IV (Col IV, 10 μg/ml) was assayed using a 6-well Transwell assay as described in Materials and Methods. Keratinocytes, untreated (black bars) or exposed to TGF-β1 for 24 hours (white bars), were seeded to the upper part of the 6-well Transwells at a density of 3×106 cells/well. After 6 hours incubation at 37°C the filters were removed and migration of cells to the lower surface of the filter was quantified. Values are the mean ± s.e.m. of triplicate samples.

Fig. 1.

Cell migration assay. Keratinocyte migration toward purified laminin 5 (LN5, 10 μg/ml), fibronectin (FN, 10 μg/ml) and collagen IV (Col IV, 10 μg/ml) was assayed using a 6-well Transwell assay as described in Materials and Methods. Keratinocytes, untreated (black bars) or exposed to TGF-β1 for 24 hours (white bars), were seeded to the upper part of the 6-well Transwells at a density of 3×106 cells/well. After 6 hours incubation at 37°C the filters were removed and migration of cells to the lower surface of the filter was quantified. Values are the mean ± s.e.m. of triplicate samples.

In order to elucidate cell behavior during the migration process, the cell adhesion properties of keratinocytes, treated for 24 hours with TGF-β1, to purified laminin 5, fibronectin and collagen IV were characterized. The adhesion of untreated cells to each of the three substrates was in accordance with that obtained previously and served as a control (Fig. 2A). Adhesion of TGF-β1-treated cells to purified laminin 5 was significantly decreased (Fig. 2B), while it was dramatically increased on fibronectin and collagen IV.

Fig. 2.

Dose-dependent cell adhesion to purified laminin 5 in comparison to fibronectin and collagen IV without (A) or with (B) a previous 24 hour treatment with 10 ng/ml/TGF-β1. Multiwell plates were coated with different concentrations of laminin 5 (•), fibronectin (△) and collagen IV (□) as indicated on the figure. Cells were added to the coated wells and the plates were incubated at 37°C for 30 minutes. For all experiments, attached cells were quantified as described in Materials and Methods. Adhesion values are mean ± s.e.m. of triplicate determinations.

Fig. 2.

Dose-dependent cell adhesion to purified laminin 5 in comparison to fibronectin and collagen IV without (A) or with (B) a previous 24 hour treatment with 10 ng/ml/TGF-β1. Multiwell plates were coated with different concentrations of laminin 5 (•), fibronectin (△) and collagen IV (□) as indicated on the figure. Cells were added to the coated wells and the plates were incubated at 37°C for 30 minutes. For all experiments, attached cells were quantified as described in Materials and Methods. Adhesion values are mean ± s.e.m. of triplicate determinations.

The unprocessed form of laminin 5 is the major component of the ECM of migratory keratinocytes

As shown by fluorescence analysis in Fig. 3A and as previously documented, laminin 5 is deposited into the matrix of cultured NHK and is present upon the substrate underneath and around the cells. As expected and shown in Fig. 3B, the TGF-β1-induced migration was accompanied by laminin 5 deposition, which when stained allowed visualization of the cell path. Therefore, the molecular form of laminin 5 present in the matrix of migrating TGF-β1 treated keratinocytes was analyzed by SDS-PAGE, and compared to that of untreated keratinocytes and purified laminin 5 (Fig. 4). After disulfide reduction, three major species were visualized by staining with Coomassie Blue in the ECM of confluent cultured keratinocytes (Fig. 4A, lane 2). These bands corresponded to those seen in the purified laminin 5 preparation (lane 1), confirming that the major component of cultured keratinocyte ECM is laminin 5. Analysis of the α? subunit by western blotting (Fig. 4B, lanes 2) revealed that most laminin 5 molecules present in normally cultured keratinocytes ECM contained the processed α3 chain (165 kDa), while faint staining of the 200 kDa form was seen. Staining of the γ2 subunit with D4B5 mAb revealed the presence of two bands, a minor band running in the position of 155 kDa and a major one in the position of 105 kDa corresponding to unprocessed and processed γ2 subunits, respectively (not shown). Indeed, nonreducing conditions (Fig. 4C) confirmed that the 400 kDa fully processed laminin 5 (α3,165; β3,140; γ2,105 kDa) was the major component of the ECM of untreated cells (lane 2). Although this form was predominant, the 460 and 440 kDa bands were also seen, confirming the presence of unprocessed laminin 5 in this ECM. As shown in lane 1 and as previously documented (Rousselle et al., 1991), purified laminin 5 contains both 440 and 400 kDa forms, corresponding to an equal mixture of the fully processed laminin 5 and the unprocessed γ2 subunit containing laminin 5. Analysis of the ECM of TGF-β1 treated keratinocytes under reducing conditions revealed the presence of three major bands running at approximately 200, 155 and 140 kDa (Fig. 4A, lane 3). Western blotting analysis of these bands revealed that the three Coomassie Blue stained bands were recognized by pAb 4101 (Fig. 4B, left panel, lane 3), suggesting that laminin 5 was expressed under different molecular forms. Blotting the reduced sample with mAb BM165 (Fig. 4B) identified the major 200 kDa band as the unprocessed α3 subunit while the 165 kDa form was minor (lane 2). Blotting reduced samples with the D4B5 mAb confirmed Coomassie Blue staining and revealed a single band running at 155 kDa, corresponding to the unprocessed γ2 chain in the matrix of migrating cells (not shown). Analysis of the material under nonreducing conditions (Fig. 4C, lane 3) revealed a major band running at the position of 460 kDa, showing that mainly unprocessed laminin 5 (α3, 200; β3, 140; γ2, 155 kDa) was found in the ECM of TGF-β1 induced migratory keratinocytes.

Fig. 3.

Immunofluorescence pattern of laminin 5 in resting and TGF-β1 induced migrating keratinocytes. The laminin ? expression pattern was examined by immunofluorescence microscopy with the pAb L132 to laminin 5 on permeabilized untreated keratinocytes (A) and keratinocytes after 24 hours treatment with TGF-β1 (B). Arrows in B show laminin 5 deposition. Bar, 50 μm.

Fig. 3.

Immunofluorescence pattern of laminin 5 in resting and TGF-β1 induced migrating keratinocytes. The laminin ? expression pattern was examined by immunofluorescence microscopy with the pAb L132 to laminin 5 on permeabilized untreated keratinocytes (A) and keratinocytes after 24 hours treatment with TGF-β1 (B). Arrows in B show laminin 5 deposition. Bar, 50 μm.

Fig. 4.

(A) Electrophoretic analysis of laminin 5 present in the ECM of resting keratinocytes compared with that of TGF-β1-induced migrating keratinocytes. Coomassie Blue staining of the reduced ECM of untreated (lane 2) and 24 hour-treated (lane 3) keratinocytes, compared with purified laminin 5 (1 μg) (lane 1), on 6% SDS-PAGE gels. Consistent with previous results, the reduced bands representing subunits of purified laminin 5 (lane 1) are identical to those found in the ECM of NHK (lane 2). Three major bands were detected in the TGF-β1 treated keratinocytes ECM (lane 3), two of which were different from those seen in lane 2. A band migrating at the 200 kDa position, and the absence of the 105 kDa processed form of the γ2 chain, suggested the presence of the unprocessed form of laminin 5 in these particular ECMs. Migration positions of the molecular mass markers are indicated to the left. (B,C) Western blot analysis of the laminin 5 subunit composition of the ECM of NHK compared with that of TGF-β1-treated keratinocytes. Analysis of the ECM of untreated (lanes 2) and 24 hour-treated (lanes 3) keratinocytes compared with purified laminin 5 (1 μg) (lanes 1) under reduced (B) and unreduced conditions (C). The ECM and purified laminin 5 were run either on a 6% SDS-PAGE gel (B) or on a 3%-5% gradient gel (C). Gels were transferred to nitrocellulose and then processed for immunoblotting using either the mAb BM165 against the α3 chain (B, right panel, C) or the pAb 4101 against all three subunits (B, left panel). Analysis of the reduced material with the pAb 4101(B) identified three major bands in the ECM of cultured keratinocytes (lane 2), corresponding to the α3, β3 and γ2 subunits and identical to those present in the purified laminin 5 preparation (lane 1). A weak band in the position of 200 kDa could also be seen. Both the 200 and 165 kDa were recognized by mAb BM165 (B, right panel, lanes 1,2). Analysis of the reduced material from TGF-β1 treated keratinocytes ECM revealed three major bands of 200, 155 and 140 kDa; the 200 and 165 kDa bands were recognized by the mAb BM165 (B, lane 3). Analysis of the unreduced material with mAb BM165 (C) revealed the presence of the previously described species of 440 and 400 kDa in the laminin 5 preparation (lane 1). Both species are found in the ECM of NHK, the 400 kDa band being predominant (lane 2). A single band of 460 kDa is stained with mAb BM165 in the ECM of TGF-β1 treated keratinocytes (lane 3).

Fig. 4.

(A) Electrophoretic analysis of laminin 5 present in the ECM of resting keratinocytes compared with that of TGF-β1-induced migrating keratinocytes. Coomassie Blue staining of the reduced ECM of untreated (lane 2) and 24 hour-treated (lane 3) keratinocytes, compared with purified laminin 5 (1 μg) (lane 1), on 6% SDS-PAGE gels. Consistent with previous results, the reduced bands representing subunits of purified laminin 5 (lane 1) are identical to those found in the ECM of NHK (lane 2). Three major bands were detected in the TGF-β1 treated keratinocytes ECM (lane 3), two of which were different from those seen in lane 2. A band migrating at the 200 kDa position, and the absence of the 105 kDa processed form of the γ2 chain, suggested the presence of the unprocessed form of laminin 5 in these particular ECMs. Migration positions of the molecular mass markers are indicated to the left. (B,C) Western blot analysis of the laminin 5 subunit composition of the ECM of NHK compared with that of TGF-β1-treated keratinocytes. Analysis of the ECM of untreated (lanes 2) and 24 hour-treated (lanes 3) keratinocytes compared with purified laminin 5 (1 μg) (lanes 1) under reduced (B) and unreduced conditions (C). The ECM and purified laminin 5 were run either on a 6% SDS-PAGE gel (B) or on a 3%-5% gradient gel (C). Gels were transferred to nitrocellulose and then processed for immunoblotting using either the mAb BM165 against the α3 chain (B, right panel, C) or the pAb 4101 against all three subunits (B, left panel). Analysis of the reduced material with the pAb 4101(B) identified three major bands in the ECM of cultured keratinocytes (lane 2), corresponding to the α3, β3 and γ2 subunits and identical to those present in the purified laminin 5 preparation (lane 1). A weak band in the position of 200 kDa could also be seen. Both the 200 and 165 kDa were recognized by mAb BM165 (B, right panel, lanes 1,2). Analysis of the reduced material from TGF-β1 treated keratinocytes ECM revealed three major bands of 200, 155 and 140 kDa; the 200 and 165 kDa bands were recognized by the mAb BM165 (B, lane 3). Analysis of the unreduced material with mAb BM165 (C) revealed the presence of the previously described species of 440 and 400 kDa in the laminin 5 preparation (lane 1). Both species are found in the ECM of NHK, the 400 kDa band being predominant (lane 2). A single band of 460 kDa is stained with mAb BM165 in the ECM of TGF-β1 treated keratinocytes (lane 3).

Identification of the α2β1 integrin as a major receptor involved in keratinocyte migration

To search for the potential receptors involved in the interactions with newly synthesized laminin 5 during cell locomotion, cell migration experiments were performed in the presence of function blocking-antibodies (Fig. 5). Fibronectin or collagen IV were used as adhesion substrates on the lower face of the Transwell chambers. First of all, inhibition of protein synthesis was performed by using cycloheximide (Fig. 5A) and induced significant reduction of migration on both fibronectin (down to 30%) and collagen IV (down to 3%). Therefore, antibodies directed against laminin 5 (Fig. 5A), as well as antibodies against the various integrin subunits, were used as described in Materials and Methods. Migration experiments conducted on fibronectin or collagen IV (Fig. 5A) revealed that pAb L132, directed against laminin 5, significantly abrogated keratinocyte migration, indicating that laminin 5 is necessary for migration. Moreover, mAb D4B5 raised against domain III of the γ2 subunit (Mizushima et al., 1998) almost totally blocked migration (down to 25%). Interestingly, mAb BM165, directed against the major laminin 5 cell binding site, enhanced cell migration (up to 150%). Control experiments conducted with function-blocking mAbs to integrin subunits (Fig. 5B,C) revealed that, as expected, mAb P1D6 against the α? integrin subunit inhibited migration on fibronectin and mAb P1E6 against the α2 integrin subunit prevented cell migration on collagen IV. Interestingly, mAb P1B5 against the α3 integrin subunit enhanced cell migration on both substrates, in a manner comparable to that obtained with mAb BM165, while mAbs directed against the α6 integrin subunit (GoH3) only weakly affected cell migration. Of particular interest was the significant inhibition of migration caused by mAb P1E6 on fibronectin (down to 25%), suggesting involvement of the α2β integrin in keratinocyte migration. In order to verify that this observation was not an artifact of the TGF-β1 treatment, the experiment was also performed with untreated NHK on fibronectin (Fig. 5C). Despite their low ability to migrate, these cells produced similar results. In view of the results obtained with P1B5 mAb, a number of control experiments were performed with other mAbs to the α3 integrin subunit (Fig. 6). The mAb ASC-1, known to block the α6β4 integrin-mediated adhesion to laminin 5 (Lichtner et al., 1998), also enhanced migration, while the nonfunction-blocking mAb A3×8 (Weitzman et al., 1993) had no effect on migration. Morover, incubating cycloheximide-pretreated cells with the P1B5 mAb prevented the cell migration enhancement that was previously obtained.

Fig. 5.

Effects of (A) inhibition of protein synthesis and antilaminin 5 antibodies and (B) function-blocking antibodies against integrin subunits on keratinocyte migration towards coated fibronectin (FN) or collagen IV (COL IV). The lower faces of 8 μm porous Transwell chambers were coated with either fibronectin (10 μg/ml) or collagen IV (10 μg/ml). After saturation with 1% BSA, TGF-β1 treated keratinocytes were seeded on the upper face of the Transwell chamber in the presence or absence of cycloheximide (25 μg/ml) or antibodies. (A) pAb L132 (serum, 1/10), mAb BM165 (12 μg/ml) or mAb D4B5 (12 μg/ml) against laminin 5, and (B) mAbs P1D6 against α? (5 μg/ml), P1B5 against α3 (5 μg/ml), GoH3 against α6 (25 μg/ml) or P1E6 against α2 (1:50), as indicated under the columns.(C)For untreated keratinocytes, antibodies of interest were tested on cell migration towards fibronectin only. After 6 hours, the extent of migration was determined as for previous figures. The results are expressed as a percentage of the control without antibodies ± s.e.m.

Fig. 5.

Effects of (A) inhibition of protein synthesis and antilaminin 5 antibodies and (B) function-blocking antibodies against integrin subunits on keratinocyte migration towards coated fibronectin (FN) or collagen IV (COL IV). The lower faces of 8 μm porous Transwell chambers were coated with either fibronectin (10 μg/ml) or collagen IV (10 μg/ml). After saturation with 1% BSA, TGF-β1 treated keratinocytes were seeded on the upper face of the Transwell chamber in the presence or absence of cycloheximide (25 μg/ml) or antibodies. (A) pAb L132 (serum, 1/10), mAb BM165 (12 μg/ml) or mAb D4B5 (12 μg/ml) against laminin 5, and (B) mAbs P1D6 against α? (5 μg/ml), P1B5 against α3 (5 μg/ml), GoH3 against α6 (25 μg/ml) or P1E6 against α2 (1:50), as indicated under the columns.(C)For untreated keratinocytes, antibodies of interest were tested on cell migration towards fibronectin only. After 6 hours, the extent of migration was determined as for previous figures. The results are expressed as a percentage of the control without antibodies ± s.e.m.

Fig. 6.

Effects of various specific antibodies against the α3 integrin subunit on keratinocyte (A) and cycloheximide-pretreated keratinocyte (B) migration towards coated fibronectin. Cell migration was studied as described in the legend to Fig. 5. TGF-β1 treated keratinocytes (A) or cycloheximide-pretreated cells (B) were seeded on the upper face of the Transwell chamber in the presence or absence of the function-blocking mAbs P1B5 (5 μg/ml) or ASC-I (5 μg/ml), or the nonfunction-blocking A3×8 (ascite, 1/100), as indicated in the figure. After 6 hours, the extent of migration was determined as for previous figures. The results are expressed as a percentage of the control without antibodies ± s.e.m.

Fig. 6.

Effects of various specific antibodies against the α3 integrin subunit on keratinocyte (A) and cycloheximide-pretreated keratinocyte (B) migration towards coated fibronectin. Cell migration was studied as described in the legend to Fig. 5. TGF-β1 treated keratinocytes (A) or cycloheximide-pretreated cells (B) were seeded on the upper face of the Transwell chamber in the presence or absence of the function-blocking mAbs P1B5 (5 μg/ml) or ASC-I (5 μg/ml), or the nonfunction-blocking A3×8 (ascite, 1/100), as indicated in the figure. After 6 hours, the extent of migration was determined as for previous figures. The results are expressed as a percentage of the control without antibodies ± s.e.m.

In view of the results obtained in the cell migration assay, analysis of the keratinocyte α2β integrin expression was performed by indirect immunofluorescence before and after migration was induced by TGF-β1. Indeed, the staining of the α2 integrin subunit under TGF-β1 treatment (Fig. 7), present in focal contacts localized at the leading edge of migrating cells, confirmed a possible involvement of this integrin in the migration process. The α2β integrins were organized into thick and well-separated aligned patches located across lamellipodia (Fig. 6Aa,b). In the case of untreated keratinocytes and in addition to strong staining at cell-cell borders, these α2 integrin-containing patches were also found in lamellipodia of cells located at the periphery of the clones (Fig. 6Ac,d). When the focus of the microscope was adjusted to the matrix located in the immediate cell environment, α2 integrin subunit staining could then be detected as light fluorescent patches attached to the substrate left behind by the migrating cells (Fig. 7B). As double immunostaining with D4B5 mAb was not feasible, study of laminin 5 expression was performed with pAb 4101 (Fig. 7C) in the same experiment. It revealed an expression pattern similar to that of α2β integrin, corresponding to the ‘left behind’ track. Analysis of the matrix beneath the cells, after removal of the cells, revealed numerous and intense footprints positively stained with the α2 integrin subunit antibody (Fig. 7G,I), which appeared to colocalize in many places with the laminin 5 staining (Fig. 7F,H). This unusual colocalization pattern with the α2 integrin subunit could not be duplicated with antibodies to other components likely to be involved in this process such as tenascin, thrombospondin, collagen XVII, NC1 domain of collagen VII (see Materials and Methods, not shown). In the case of untreated clones, the leftover α2 integrin subunit staining remained at cell-cell borders (Fig. 7E). Concomitant analysis of α6 and α3 integrin subunits under TGF-β1 treatment also revealed, for both integrin subunits, a staining similar to that of laminin 5 (not shown).

Fig. 7.

Immunofluorescence pattern of the α2 integrin subunit in TGF-β1 induced migrating keratinocytes. The α2 integrin expression pattern was examined by immunofluorescence microscopy on permeabilized keratinocytes treated for 24 hours with TGF-β1 (Aa,b, B) and untreated keratinocytes (Ac,d) with mAb P1E6. In the case of double staining with laminin 5 (C), cells were incubated with pAb 4101. In a second set of experiments (D-I), untreated (D,E) and TGF-β1-treated (F-I) cells were removed prior to immunostaining with pAb 4101 to laminin 5 (D,F,H) and mAb P1E6 (E,G,I). The α2 staining is present at the cell-cell junction in the case of untreated cell ECM (E), while it appears as numerous intense footprints (G,I) colocalizing with the laminin 5 staining (F,H) in the case of TGF-β1 treated keratinocyte ECM. Bars, 10 μm (A-C, H-I); 20 μm (D-G).

Fig. 7.

Immunofluorescence pattern of the α2 integrin subunit in TGF-β1 induced migrating keratinocytes. The α2 integrin expression pattern was examined by immunofluorescence microscopy on permeabilized keratinocytes treated for 24 hours with TGF-β1 (Aa,b, B) and untreated keratinocytes (Ac,d) with mAb P1E6. In the case of double staining with laminin 5 (C), cells were incubated with pAb 4101. In a second set of experiments (D-I), untreated (D,E) and TGF-β1-treated (F-I) cells were removed prior to immunostaining with pAb 4101 to laminin 5 (D,F,H) and mAb P1E6 (E,G,I). The α2 staining is present at the cell-cell junction in the case of untreated cell ECM (E), while it appears as numerous intense footprints (G,I) colocalizing with the laminin 5 staining (F,H) in the case of TGF-β1 treated keratinocyte ECM. Bars, 10 μm (A-C, H-I); 20 μm (D-G).

The short arm of the γ2 subunit is the ligand for the α2β1 integrin

As keratinocyte migration appeared to involve an α2β integrin interaction with laminin 5, and as the antilaminin γ2 subunit mAb D4B5 interfered with cell migration in a manner comparable to that of mAb P1E6, further experiments were performed in order to verify that these two phenomena were related. To test the potential involvement of the α2β integrin binding to the short arm of the laminin γ2 chain, solid-phase assay binding studies were performed using a soluble integrin preparation obtained from a keratinocyte lysate interacting with immobilized laminin 5 (Fig. 8). Since purified laminin 5 preparation is composed of both processed and unprocessed γ2-containing laminin 5 molecules (Fig. 4C), such substrate was used in this experiment. To confirm previous studies showing the direct interaction between α2β integrin and laminin 5 (Orian-Rousseau et al., 1998), the binding of α2β integrin to immobilized laminin 5 was measured in comparison to laminin 1 (Fig. 8A). The specificity of both molecular interactions was verified by the complete inhibition obtained with mAb P1E6 (Fig. 8B). Incubation of the immobilized ligand with mAb D4B5, against domain III of the laminin γ2 chain prior to the interaction, totally prevented binding of the α2β integrin to laminin 5, while mAb 6F12 against the laminin β3 chain did not. None of these mAbs affected the binding to laminin 1 (Fig. 8B).

Fig. 8.

α2β integrin binding to immobilized laminin 5 (LN-5; 40 μg/ml) and laminin 1 (LN-1; 20 μg/ml) determined by solid-phase assays as described under Materials and Methods. (B) Inhibition of α2β integrin binding to laminin 5 ligand (40 μg/ml) by mAb P1E6 against the α2 integrin subunit and mAb D4B5 against domain III located within the short arm of the laminin γ2 subunit. mAb 6F12 against the laminin β3 subunit was used as an internal control. The control experiment shows that α2β integrin binding to laminin 1 (20 μg/ml) is blocked by mAb P1E6, but not by mAbs D4B5 and 6F12. The level of binding in the absence of antibody was set to 100%.

Fig. 8.

α2β integrin binding to immobilized laminin 5 (LN-5; 40 μg/ml) and laminin 1 (LN-1; 20 μg/ml) determined by solid-phase assays as described under Materials and Methods. (B) Inhibition of α2β integrin binding to laminin 5 ligand (40 μg/ml) by mAb P1E6 against the α2 integrin subunit and mAb D4B5 against domain III located within the short arm of the laminin γ2 subunit. mAb 6F12 against the laminin β3 subunit was used as an internal control. The control experiment shows that α2β integrin binding to laminin 1 (20 μg/ml) is blocked by mAb P1E6, but not by mAbs D4B5 and 6F12. The level of binding in the absence of antibody was set to 100%.

The α2β1 integrin plays a role in cell spreading on laminin 5

To gain insight into the possible involvement of the α2β integrin in cell adhesion to laminin 5 during cell migration, cell adhesion experiments using normal keratinocytes and TGF-β1 induced migrating keratinocytes were carried out with anti-integrin-specific antibodies including mAb P1E6 and D4B5 (Fig. 9). In a manner comparable to that obtained previously with NHK (Rousselle and Aumailley, 1994), adhesion of TGF-β1 treated keratinocytes to laminin 5 was totally abrogated by mAbs BM165, P4C10 and P1B5 while GoH3 had no effect (Fig. 9A). Preincubation of the substrate with mAb D4B5 prior to the cell adhesion experiment was not inhibitory and use of mAb P1E6 produced a substantial disturbance of cell adhesion as compared to the control. Analysis of cell spreading by phase-contrast microscopy and measurement of cell diameters (Fig. 9B) revealed that mAb P1E6, and D4B5 to a lesser extent, was inhibitory of cell spreading rather than cell adhesion itself. The effect of both mAbs on cell spreading on laminin 5 was minimal in the case of untreated keratinocytes. Indeed, mAb P1E6 impeded cell spreading either on laminin 5 or on the ECM of TGF-β1-treated keratinocytes (not shown) as compared to the controls without antibodies.

Fig. 9.

Quantitative and qualitative analysis of cell adhesion to purified laminin 5 in the presence of various anti-laminin 5 and anti-integrin mAbs, including P1E6. TGF-β1-treated or untreated keratinocytes were seeded on purified laminin 5 (10 μg/ml) after pre-incubation with mAbs P4C10 (1:50), GoH3 (1:50), P1B5 (1:200) and P1E6 (1:50). In the case of anti-laminin 5 mAbs, the coated wells were incubated with BM165 (10 μg/ml) or D4B5 (10 μg/ml) 60 minutes before the experiment. After cell adhesion assays, the extent of adhesion was measured and expressed as the percentage of adhesion in the absence of antibodies. For spreading analysis, micrographs were taken under phase-contrast microscopy and, for each case, cell sizes were determined by measurement of largest diameter of 50 cells on enlarged pictures.

Fig. 9.

Quantitative and qualitative analysis of cell adhesion to purified laminin 5 in the presence of various anti-laminin 5 and anti-integrin mAbs, including P1E6. TGF-β1-treated or untreated keratinocytes were seeded on purified laminin 5 (10 μg/ml) after pre-incubation with mAbs P4C10 (1:50), GoH3 (1:50), P1B5 (1:200) and P1E6 (1:50). In the case of anti-laminin 5 mAbs, the coated wells were incubated with BM165 (10 μg/ml) or D4B5 (10 μg/ml) 60 minutes before the experiment. After cell adhesion assays, the extent of adhesion was measured and expressed as the percentage of adhesion in the absence of antibodies. For spreading analysis, micrographs were taken under phase-contrast microscopy and, for each case, cell sizes were determined by measurement of largest diameter of 50 cells on enlarged pictures.

Keratinocyte α2β1 integrin expression at the migrating front during wound closure

To understand better the possible involvement of the α2β?integrin in keratinocyte migration in vivo, human cutaneous wound biopsies from a full-thickness burn patient were assessed by immunolabeling of laminin 5 and various integrin subunits. Our findings were in accordance with earlier studies demonstrating an increase of laminin 5 expression and deposition by cells located at the regenerating epidermal margin of skin wounds (Larjava et al., 1993; Ryan et al., 1994; Kainulainen et al., 1998) (not shown). Analysis of integrin subunits revealed that migrating cells present at the leading edge stained brightly with mAb P1E6 (Fig. 10B), suggesting that these cells were actively expressing the α2β integrin possibly involved in cell-matrix interactions. Staining domain III of the laminin γ2 subunit with D4B5 mAb on serial section revealed a similar localization (Fig. 10C). Observation at higher magnification shows strong intracellular and extracellular staining at the migrating front, for both antigens (Fig. 10E,F).

Fig. 10.

Immunolocalization of laminin 5 γ2 epitope and integrin α2β in regenerating epidermis. Immunostaining, with Trypan Blue staining of nuclei (A,D), mAbs P1E6 (B,E), D4B5 (C,F) in a 5-day-old full-thickness human skin wound. Staining of nuclei (A,D), allows visualization of epidermis (white E) and dermis (white D) at the leading edge of reepithelialization (white arrows). Staining of the α2β integrin is seen at the leading edge (B) similar to that obtained with staining of the laminin 5 γ2 chain (C). Analysis of the cells (D) and α2 integrin subunit staining at higher magnification (E) reveals an increased and reorganized expression of α2β integrins on cells localized at the wound margin of the regenerating epidermis (white arrowheads) as compared to the typical cell-cell junction location in resting epidermis (not shown). While, in normal conditions, laminin 5 localizes to the basement membrane beneath keratinocytes (bm), immunostaining of the wound margin reveals the presence of the laminin 5 γ2 chain at the intracellular level and in the entire newly formed basement membrane deposited under the epithelium (F). Bars, 50 μm.

Fig. 10.

Immunolocalization of laminin 5 γ2 epitope and integrin α2β in regenerating epidermis. Immunostaining, with Trypan Blue staining of nuclei (A,D), mAbs P1E6 (B,E), D4B5 (C,F) in a 5-day-old full-thickness human skin wound. Staining of nuclei (A,D), allows visualization of epidermis (white E) and dermis (white D) at the leading edge of reepithelialization (white arrows). Staining of the α2β integrin is seen at the leading edge (B) similar to that obtained with staining of the laminin 5 γ2 chain (C). Analysis of the cells (D) and α2 integrin subunit staining at higher magnification (E) reveals an increased and reorganized expression of α2β integrins on cells localized at the wound margin of the regenerating epidermis (white arrowheads) as compared to the typical cell-cell junction location in resting epidermis (not shown). While, in normal conditions, laminin 5 localizes to the basement membrane beneath keratinocytes (bm), immunostaining of the wound margin reveals the presence of the laminin 5 γ2 chain at the intracellular level and in the entire newly formed basement membrane deposited under the epithelium (F). Bars, 50 μm.

In this study, we have analyzed the role of laminin 5, newly synthesized and secreted by migrating keratinocytes. Since NHK do not tend to move in normal culture conditions, but on the contrary, can form stable hemidesmosomal-like structures, we have induced them to migrate by exposure to TGF-β1. Moreover, since TGF-β1 prevents NHK proliferation (reviewed in Moses et al., 1990), this important feature permitted the study to be focused on cell migration without interference from cell proliferation. In agreement with studies conducted with other epithelial cells (Miettinen et al., 1994; Boland et al., 1996), the data presented here demonstrate that TGF-β1 induced significant increase in NHK migration when cells were placed on either fibronectin or collagen IV. Absence of migration on purified laminin 5, whether cells were TGF-β1-treated or not, confirms previous studies showing that processed-α? containing laminin 5 does not support migration of keratinocytes or other epithelial cell lines such as SCC12 and MCF10A (O’Toole et al., 1997; Giannelli et al., 1997; Goldfinger et al., 1998). In contrast, this substrate has been shown to support migration of a number of cell lines including glioma or HT1080 fibrosarcoma cells (Fukushima et al., 1998; Hirosaki et al., 2000; Koshikawa et al., 2000). Recent data indicates that the ability for these particular cells to constitutively migrate on laminin 5 is linked to their capability to specifically express the plasma-membrane-bound MT1-MMP metalloprotease and cleave the γ2 chain to an 80 kDa peptide (Koshikawa et al., 2000). This mechanism is likely to be similar to that observed in scattering of these cells by the addition of soluble laminin 5 (Miyasaki et al., 1993; Kikkawa et al., 1994; Koshikawa et al., 2000). Adding soluble laminin 5 to NHK does not have a scattering effect (O’Toole et al., 1997). On the contrary, it was shown to inhibit collagen I-driven motility migration (O’Toole et al., 1997). It is likely that this process is strictly dependent on the cell type and that keratinocyte migration may require a different type of interaction with laminin 5.

Our cell adhesion studies, showing dramatic enhancement of cell adhesion to fibronectin and collagen IV with TGF-β1 treatment, reveal a change in integrin expression similar to that previously documented (Sheppard et al., 1992; Gailit et al., 1994). Of novel interest are our cell adhesion data reporting a significant decrease in cell adhesion to purified laminin 5 (down to 40%) when cells were forced to migrate, suggesting an absolute necessity for the cells to decrease their adhesion to processed laminin 5 in order to migrate. In addition, our results corroborate previous observations describing a TGF-β1 induced decrease in α6β4 integrin expression in various human cell lines (Heino and Massague, 1989; Kumar et al., 1995) and in keratinocytes (Tennenbaum et al., 1996; Zambruno et al., 1995). Despite its decreased adhesion-promoting activity for TGF-β1-induced migrating keratinocytes, laminin 5 synthesis has, however, been shown to be upregulated by TGF-β1 (Korang et al., 1995), and more generally so during the in vivo epidermal cell migration processes (Larjava et al., 1993; Ryan et al., 1994; Kainulainen et al., 1998; Salo et al., 1999). Moreover, endogenous laminin 5 has been proposed to play a crucial role in keratinocyte migration in vitro (Zhang and Kramer, 1996). Our immunohistochemical and electrophoretic analysis of the ECM beneath TGF-β1 induced migrating cells revealed that its main component is laminin 5. Under these conditions, we demonstrate that laminin 5 is mainly present in its unprocessed form of 460 kDa, suggesting that processing of both α? and γ2 chain is prevented. In contrast, the predominant molecular form of laminin 5 present in the matrix of confluent keratinocytes is the fully processed form, confirming that, in normal conditions, laminin 5 is rapidly processed after secretion. In the case of NHK, various amounts of the unprocessed form were found, depending on the level of cell confluency (P. Rousselle, unpublished observation). Our observations are in agreement with a recent report showing a migratory promoting activity of unprocessed α?-containing laminin 5 (Goldfinger et al., 1998). In addition, our results suggest that the lack of cleavage of the γ2 subunit may also be associated with keratinocyte migration.

Our cell migration inhibition experiment after either cycloheximide treatment or use of the pAb L132 against laminin 5, showed that laminin 5 deposition appears to be critical for cell migration. Of particular interest, our results showed enhancement of migration when cells were incubated with the mAb P1B5 directed against the α3 integrin subunit. The use of another mAb known to block α6β4 integrin adhesion to laminin 5 (ASC-1, Lichtner et al., 1998) also induced migration enhancement, indicating that the α6β4 integrin does not promote migration but, on the contrary, it impedes this process and renders the cells more adherent. Our results showed absence of migration enhancement with the noninhibitory mAb A3×8 (Weitzman et al., 1993), as well as the absence of migration enhancement by the P1B5 mAb when cells were pretreated with cyclohemide. These data strongly suggest that enhanced migration is a result of an inhibition of the α6β4 integrin adhesion to laminin 5 rather than an activation caused by the P1B5 antibody. Similar observations were reported in previous studies conducted with lung carcinoma cells (Barr et al., 1998) and NHK (Kim et al., 1992; O’Toole et al., 1997), showing that a decrease in α6β4 integrin expression or a blocking of its function was associated with an increased cell migration ability. In vivo findings support this hypothesis by showing that a reduction of α6β4 is associated with the increased virulence of epithelial cancers (Adachi et al., 1998; Bartolazzi et al., 1995; Koukoulis et al., 1991). In accordance with O’Toole et al., 1998, our results show that preincubation of the cells with mAb BM165 resulted in an enhancement of their migratory capacity in a manner comparable to that obtained with the mAb P1B5. These data strengthen the hypothesis that the mAb BM165 epitope on laminin 5 is involved in stable cell adhesion and maintenance of HDs. As interactions with both α6β4 and α6β4 integrins are thought to occur with sites on laminin 5 corresponding to the mAb BM165 epitope (Rousselle and Aumailley, 1994), it was not clear which integrin was blocked when mAb BM165 was used. Since function-blocking antibodies to the α6 integrin subunits produced weak perturbation of cell migration, we propose that the α6β4 interaction with newly synthesized laminin 5 strongly impedes keratinocyte migration. The latter suggestion is reinforced by recent work from Sterk and coworkers, suggesting that α6β4 integrin-CD151 clusters may precede and facilitate the formation of HDs (Sterk et al., 2000). However, our findings contradict data presented by others, suggesting a crucial role for the interaction between the α6β4 integrin and the α? chain of laminin 5 during migration (Zhang and Kramer, 1997; Goldfinger et al., 1999). It is conceivable that, in the vitro wound-closure models presented in both studies, antibodies to either laminin 5 or α6β4 integrins inhibited cell migration by interfering with cell adhesion. Since laminin 5 is the main component synthesized and deposited by migrating cells, absence of a provisional matrix or another substrate for the cells to attach to may be the reason for the observed inhibition. This may also be the case in the experiments in which authors performed their cell migration assays on immobilized laminin 5 (Fukushima et al., 1998).

In accordance with our results, in vitro studies conducted with α6β4 knock-out keratinocytes indicate that the α6β4 integrin may trans-dominantly inhibit fibronectin and collagen IV receptor functions, confirming that α6β4 integrin does not facilitate migration processes on these substrates (Hodivala-Dilke et al., 1998). Moreover, data obtained using breast carcinoma cell lines propose a mechanism involving a possible α6β4 integrin-negative trans-regulation of the α2β integrin (Lichter et al., 1998). Furthermore, there is growing evidence suggesting that the α6β4 integrin may have a role to play in ECM assembly by establishing and/or maintaining integrity of the basement membrane (DiPersio et al., 1997; Wu et al., 1995). However, and as our model is restricted to the study of migrating cells in the Transwell chambers system only, we cannot exclude the possibility that the α6β4 integrin may actively participate in the re-epithelialization process by mediating different and cooperative signaling pathways.

Interestingly, our results demonstrate a role for the α2β integrin during keratinocyte migration whether the cells are TGF-β1 treated or not. Our experiments showing inhibition of keratinocyte migration by mAb D4B5 directed against domain III of the γ2 subunit (Mizushima et al., 1998), in a manner comparable to that obtained with mAb P1E6 against the α2 integrin subunit, were a primary indication of a possible relationship between these two events. A crucial role for the γ2 subunit in cell migration was also proposed in a study showing that a pAb to domain III was able to block cell migration of mouse keratinocytes (Salo et al., 1999). Other studies reported a mechanism by which a specific and unique cleavage of the γ2 subunit by matrix metalloprotease-2 (MMP2), or Membrane Type1-MMP, induced migration of various epithelial cell types on purified laminin 5 (Giannelli et al., 1997; Koshikawa et al., 2000). The mechanism occurring in our experiments is likely to be different since we never detected the migration-related 80 kDa cleaved γ2 fragment in the ECM of TGFβ1-treated migrating keratinocytes. Furthermore, our immunolabelling experiments showing perfect colocalization of the α2β integrin with laminin 5 in tracks left behind by migrating cells, strongly suggested that the α2β integrin was likely to bind the unprocessed form of laminin 5. We also detected α2β integrin-containing-focal contacts in peripheral lamellipodia of untreated cells located at the edges of growing clones. Laminin 5 was shown to be actively synthesized by growing keratinocytes (Rousselle et al., 1991) and recent data reported identification of the unprocessed α? chain at the periphery of growing epithelial clones (Goldfinger et al., 1999). Since processing of the γ2 chain was shown to occur at a slower rate than that of the α? chain (Marinkovich et al., 1992), it is conceivable that the γ2 chain may also be unprocessed in this precise location. This would suggest that the α2β integrin may transiently interact with unprocessed laminin 5 after its secretion. Previous studies (Orian-Rousseau et al., 1998) reported an interaction between laminin 5 and purified α2β integrin. Our solid-phase interaction studies demonstrating that solubilized α2β integrin binding to purified laminin 5 is inhibited by mAb D4B5, show for the first time that the α2β integrin binds to the short arm of the γ2 subunit in a specific manner.

The α2β integrins were originally identified as major receptors for several collagen types (Elices and Hemler, 1989; Languino et al., 1989; Kirchhofer et al., 1990), but they have also been shown to bind to laminin 1, although with lower efficiency (Pfaff et al., 1994). Integrin α2β binding to laminin 1 was originally mapped to fragment E1XNd, corresponding to the N-terminal portion of the molecule (Pfaff et al., 1994). Recent studies proposed an interaction with the N-terminal globular domain VI of the laminin α? chain (Ettner et al., 1998) as well as of the laminin α2 chain (Colognato et al., 1997). Our results suggest that binding of the α2β integrin occurs within the γ2 chain of laminin 5. In that regard, recent work reported identification of an α2β integrin binding sequence within domain IV of the γ1 subunit in a region with high homology with the γ2 subunit (Nomizu et al., 1997). We can speculate that the α2β integrin binding site on the γ2 chain may be localized within the N-terminal portion, which is removed after proteolytic cleavage during maturation of the molecule (Vailly et al., 1994; Amano et al., 2000). This hypothesis is strengthened by our results showing loss of the α2β integrin colocalization with processed laminin 5 in the ECM confluent keratinocytes (Fig. 7D,E). However, we cannot confirm this suggestion since the mAb D4B5 epitope is localized within the portion of domain III that remains on the γ2 subunit after cleavage (Mizushima et al., 1998). Inhibition of migration could then be explained by steric hindrance caused by the D4B5 mAb. Further studies are currently being conducted to resolve this question.

Other studies have proposed a role for the α2β integrin in the migration process, with absence of an obvious adhesive function (Lochter et al., 1999), suggesting a role for these integrin-ECM interactions in the regulation of matrix metalloproteinase expression. Our adhesion experiments propose a role for the α2β integrin in keratinocytes spreading on purified laminin 5. Indeed we have shown that incubation of TGF-β1 treated keratinocytes with mAb P1E6 prior to cell adhesion to purified laminin 5 or unprocessed laminin 5-containing matrices did not affect adhesion but strongly impeded spreading. Why the function of the α2β integrin in keratinocytes spreading on laminin 5 is seen only when the cells have been exposed to TGF-β1 remains to be elucidated. As previously suggested (Hodivala-Dilke et al., 1998; Lichtner et al., 1998), the α6β4 integrin may trans-dominantly restrict α2β integrin function, and it is conceivable that the downregulated expression of the α6β4 integrin by TGF-β1 may allow the α2β integrin to interact with the laminin γ2 chain. We can speculate that, when cells become stationary, the involvement of the α2β integrin in migration may be abolished upon re-expression of the α6β4 integrin.

The crucial participation of the α2β integrin was shown for initiation of keratinocyte migration over dermal type I collagen (Pilcher et al., 1997). A recent study using a similar model has provided evidence that keratinocytes deposit laminin 5 while migrating over collagen I, and that two different cell populations can be distinguished at the wound margin, depending on their α2β integrin-mediated binding to collagen I or their α6β4 integrin-mediated binding to deposits of laminin 5 (Nguyen et al., 2000). While our approach does not allow us to assign our findings to a particular cell population, we consider the possibility that α2β integrin binding to laminin 5 may represent a secondary event occurring after migration has been induced.

The strong pericellular expression of the α2β integrin in migrating keratinocytes during the process of wound healing, correlated to the staining we observed with mAb D4B5, indicates that the transient interaction of α2β integrin with the laminin γ2 chain may be of physiological relevance. In summary, the present study indicates that, among integrins involved in laminin 5 interactions, α2β seems to have a role to play during migration of keratinocytes while α6β4 retards it. We show that the α2β integrin binds to the short arm of the γ2 chain. In addition to controlling cell growth, TGFβ certainly has an important role to play in epidermal repair by regulating the transcription of laminin 5 and by controlling its interactions with keratinocytes by modulating the integrin expression at the cell surface.

We are indebted to Prof. Burgeson for the generous gift of 6F12 and NP32 mAbs, Dr Hemler for A3×8 mAb, Dr Meneguzi for pAb L 799P, Dr Clezardin for pAb Ab-8, Prof. Aumailley for the kind gift of laminin 1 and to Dr Braye for providing us with the wound biopsy. We thank M. Gonzalez for excellent technical assistance and A. Bosch for artwork. This work was supported by grants from the CNRS, the Association pour la Recherche sur le Cancer, the Ligue Nationale contre le Cancer (Rhône-Alpes, Loire), a CNRS-INSERM grant (Adhésion Cellules/Biomatériaux) and support as JSID’s International Fellowship Shiseido Award to P. Rousselle. F. Décline was a recipient of a studentship from the French government and the Fondation pour la Recherche Médicale.

Adachi
,
M.
,
Taki
,
T.
,
Huang
,
C.
,
Higashiyama
,
M.
,
Doi
,
O.
,
Tsuji
,
T.
and
Miyake
,
M.
(
1998
).
Reduced integrin alpha3 expression as a factor of poor prognosis of patients with adenocarcinoma of the lung
.
J. Clin. Oncol
.
16
,
1060
1067
.
Adams
,
J. C.
and
Watt
,
F. M.
(
1990
).
Changes in keratinocyte adhesion during terminal differentiation: reduction in fibronectin binding precedes alpha 5 beta 1 integrin loss from the cell surface
.
Cell
63
,
425
435
.
Amano
,
S.
,
Scott
,
I. C.
,
Takahara
,
K.
,
Koch
,
M.
,
Champliaud
,
M. F.
,
Gerecke
,
D. R.
,
Keene
,
D. R.
,
Hudson
,
D. L.
,
Nishiyama
,
T.
,
Lee
,
S.
,
Greenspan
,
D. S.
and
Burgeson
R. E.
(
2000
).
Bone morphogenetic protein 1 is an extracellular processing enzyme of the laminin 5 gamma 2 chain
.
J. Biol. Chem
.
275
,
22728
22735
.
Baker
,
S. E.
,
Hopkinson
S. B.
,
Fitchmun
,
M.
,
Andreason
,
G. L.
,
Frasier
,
F.
,
Plopper
,
G.
,
Quaranta
,
V.
and
Jones
,
J. C.
(
1996
).
Laminin-5 and hemidesmosomes: role of the alpha 3 chain subunit in hemidesmosome stability and assembly
.
J. Cell Sci
.
109
,
2509
2520
.
Barr
,
L. F.
,
Campbell
,
S. E.
,
Bochner
,
B. S.
and
Dang
,
C. V.
(
1998
).
Association of the decreased expression of alpha3 beta1 integrin with the altered cell: environmental interactions and enhanced soft agar cloning ability of c-myc-overexpressing small cell lung cancer cells
.
Cancer Res
.
58
,
5537
5545
.
Bartolazzi
,
A.
,
Cerboni
,
C.
,
Flamini
,
G.
,
Bigotti
,
A.
,
Lauriola
,
L.
and
Natali
,
P. G.
(
1995
).
Expression of alpha 3 beta 1 integrin receptor and its ligands in human lung tumors
.
Int. J. Cancer
64
,
248
252
.
Boland
,
S.
,
Boisvieux-Ulrich
,
E.
,
Houcine
,
O.
,
Baeza-Squiban
,
A.
,
Pouchelet
,
M.
,
Schoevaert
,
D.
and
Marano
,
F.
(
1996
).
TGF beta 1 promotes actin cytoskeleton reorganization and migratory phenotype in epithelial tracheal cells in primary culture
.
J. Cell Sci
.
109
,
2207
2219
.
Carter
,
W. G.
,
Kaur
,
P.
,
Gil
,
S. G.
,
Gahr
,
P. J.
and
Wayner
,
E. A.
(
1990a
).
Distinct functions for integrins alpha 3 beta 1 in focal adhesions and alpha 6 beta 4/bullous pemphigoid antigen in a new stable anchoring contact (SAC) of keratinocytes: relation to hemidesmosomes
.
J. Cell Biol
.
111
,
3141
3154
.
Carter
,
W. G.
,
Ryan
,
M. C.
and
Gahr
,
P. J.
(
1991
).
Epiligrin, a new cell adhesion ligand for integrin alpha 3 beta 1 in epithelial basement membranes
.
Cell
65
,
599
610
.
Carter
,
W. G.
,
Wayner
,
E. A.
,
Bouchard
,
T. S.
and
Kaur
,
P.
(
1990b
).
The role of integrins alpha 2 beta 1 and alpha 3 beta 1 in cell-cell and cell-substrate adhesion of human epidermal cells
.
J. Cell Biol
.
110
,
1387
1404
.
Cavani
,
A.
,
Zambruno
,
G.
,
Marconi
,
A.
,
Manca
,
V.
,
Marchetti
,
M.
and
Giannetti
,
A.
(
1993
).
Distinctive integrin expression in the newly forming epidermis during wound healing in humans
.
J. Invest. Dermatol
.
101
,
600
604
.
Clark
,
R. A. F.
(
1996
).
Wound repair: overview and general considerations
.
In The Molecular and Cellular Biology of Wound Repair
(ed.
R. A. F.
Clark
), pp.
3
50
.
New York
:
Plenum Press Co
.
Colognato
H.
,
Mac Carrick
,
M.
,
O’Rear
,
J. J.
and
Yurchenco
,
P. D.
(
1997
).
The laminin alpha2 chain short arm mediates cell adhesion through both the alpha1beta1 and alpha2beta1 integrins
.
J. Biol. Chem
.
272
,
29330
29336
.
DiPersio
,
C. M.
,
Hodivala-Dilke
,
K. M.
,
Jaenisch
,
R.
,
Kreidberg
,
J. A.
and
Hynes
,
R. O.
(
1997
).
alpha3beta1 integrin is required for normal development of the epidermal basement membrane
.
J. Cell Biol
.
137
,
729
742
.
Elices
,
M. J.
and
Hemler
,
M. E.
(
1989
).
The human integrin VLA-2 is a collagen receptor on some cells and a collagen/laminin receptor on others
.
Proc. Natl. Acad. Sci. USA
86
,
9906
9910
.
Ettner
,
N.
,
Gohring
,
W.
,
Sasaki
,
T.
,
Mann
,
K.
and
Timpl
,
R.
(
1998
).
The N-terminal globular domain of the laminin alpha1 chain binds to alpha1beta1 and alpha2beta1 integrins and to the heparan sulfate-containing domains of perlecan
.
FEBS Lett
.
430
,
217
21
.
Fukushima
.
Y.
,
Ohnishi
,
T.
,
Arita
,
N.
,
Hayakawa
,
T.
and
Sekiguchi
,
K.
(
1998
).
Integrin alpha3beta1-mediated interaction with laminin-5 stimulates adhesion, migration and invasion of malignant glioma cells
.
Int. J. Cancer
76
,
63
72
.
Gailit
,
J.
,
Welch
,
M. P.
and
Clark
,
R. A.
(
1994
).
TGF-beta 1 stimulates expression of keratinocyte integrins during re-epithelialization of cutaneous wounds
.
J. Invest. Dermatol
.
103
,
221
227
.
Giannelli
,
G.
,
Falk-Marzillier
,
J.
,
Schiraldi
,
O.
,
Stetler-Stevenson
,
W. G.
and
Quaranta
,
V.
(
1997
).
Induction of cell migration by matrix metalloprotease-2 cleavage of laminin-5
.
Science
277
,
225
228
.
Goldfinger
,
L. E.
,
Hopkinson
,
S. B.
,
deHart
G. W.
,
Collawn
,
S.
,
Couchman
,
J. R.
and
Jones
.
J. C.
(
1999
).
The alpha3 laminin subunit, alpha6beta4 and alpha3beta1 integrin coordinately regulate wound healing in cultured epithelial cells and in the skin
.
J. Cell Sci
.
112
,
2615
2629
.
Goldfinger
,
L. E.
,
Stack
M. S.
and
Jones
,
J. C.
(
1998
).
Processing of laminin-5 and its functional consequences: role of plasmin and tissue-type plasminogen activator
.
J. Cell Biol
.
141
,
255
265
.
Gospodarowicz
,
D.
(
1984
).
Preparation of extracellular matrices produced by cultured bovine corneal endothelial cells and PF-HR-9 endodermal cells: their use in cell culture
.
In Methods for Preparation of Media, Supplements, and Substrata for Serum-free Animal Cell Culture
, pp.
275
293
.
New York
:
A. R. Liss Inc
.
Heino
,
J.
and
Massague
,
J.
(
1989
).
Transforming growth factor-beta switches the pattern of integrins expressed in MG-63 human osteosarcoma cells and causes a selective loss of cell adhesion to laminin
.
J. Biol. Chem
.
264
,
21806
21811
.
Hertle
,
M. D.
,
Adams
J. C.
and
Watt
,
F. M.
(
1991
).
Integrin expression during human epidermal development in vivo and in vitro
.
Development
112
,
193
206
.
Hirosaki
,
T.
,
Mizushima
,
H.
,
Tsubota
,
Y.
,
Moriyama
,
K.
and
Miyazaki
,
K.
(
2000
).
Structural requirement of carboxyl-terminal globular domains of laminin alpha 3 chain for promotion of rapid cell adhesion and migration by laminin-5
.
J. Biol. Chem
.
275
,
22495
502
.
Hodivala-Dilke
,
K. M.
,
DiPersio
C. M.
,
Kreidberg
,
J. A.
and
Hynes
,
R. O.
(
1998
).
Novel roles for alpha3beta1 integrin as a regulator of cytoskeletal assembly and as a trans-dominant inhibitor of integrin receptor function in mouse keratinocytes
.
J. Cell Biol
.
142
,
1357
1369
.
Humphries
,
M. J.
(
1991
).
Wound Healing
.
In Receptors for Extracellular Matrix
(ed.
J. A.
Mc Donald
and
R. P.
Macham
), pp.
220
253
.
San Diego
:
Academic Press
.
Juhasz
,
I.
,
Murphy
,
G. F.
,
Yan
,
H. C.
,
Herlyn
,
M.
and
Albelda
,
S. M.
(
1993
).
Regulation of extracellular matrix proteins and integrin cell substratum adhesion receptors on epithelium during cutaneous human wound healing in vivo
.
Am. J. Pathol
.
143
,
1458
69
.
Kainulainen
,
T.
,
Hakkinen
,
L.
,
Hamidi
,
S.
,
Larjava
,
K.
,
Kallioinen
,
M.
,
Peltonen
,
J.
,
Salo
,
T.
,
Larjava
,
H.
and
Oikarinen
,
A.
(
1998
).
Laminin-5 expression is independent of the injury and the microenvironment during reepithelialization of wounds
.
J. Histochem. Cytochem
.
46
,
353
360
.
Kaufmann
,
R.
,
Frosch
,
D.
,
Westphal
,
C.
,
Weber
,
L.
and
Klein
,
C. E.
(
1989
).
Integrin VLA-3: ultrastructural localization at cell-cell contact sites of human cell cultures
.
J. Cell Biol
.
109
,
1807
1815
.
Kikkawa
,
Y.
,
Umeda
,
M.
and
Miyazaki
,
K.
(
1994
).
Marked stimulation of cell adhesion and motility by ladsin, a laminin-like scatter factor
.
J. Biochem
.
116
,
862
869
.
Kim
,
J. P.
,
Zhang
,
K.
,
Kramer
,
R. H.
,
Schall
,
T. J.
and
Woodley
,
D. T.
(
1992
).
Integrin receptors and RGD sequences in human keratinocyte migration: unique anti-migratory function of alpha 3 beta 1 epiligrin receptor
.
J. Invest. Dermatol
.
98
,
764
970
.
Kirchhofer
,
D.
,
Languino
,
L. R.
,
Ruoslahti
,
E.
and
Pierschbacher
,
M. D.
(
1990
).
Alpha 2 beta 1 integrins from different cell types show different binding specificities
.
J. Biol. Chem
.
265
,
615
618
.
Koshikawa
,
N.
,
Giannelli
,
G.
,
Cirulli
,
V.
,
Miyazaki
,
K.
and
Quaranta
,
V.
(
2000
).
Role of cell surface metalloprotease MT1-MMP in epithelial cell migration over laminin-5
.
J. Cell Biol
.
148
,
615
624
.
Korang
,
K.
,
Christiano
,
A. M.
,
Uitto
,
J.
and
Mauviel
,
A.
(
1995
).
Differential cytokine modulation of the genes LAMA3, LAMB3, and LAMC2, encoding the constitutive polypeptides, alpha 3, beta 3, and gamma 2, of human laminin 5 in epidermal keratinocytes
.
FEBS Lett
.
368
,
556
558
.
Koukoulis
,
G. K.
,
Virtanen
,
I.
,
Korhonen
,
M.
,
Laitinen
,
L.
,
Quaranta
,
V.
and
Gould
,
V. E.
(
1991
).
Immunohistochemical localization of integrins in the normal, hyperplastic, and neoplastic breast. Correlations with their functions as receptors and cell adhesion molecules
.
Am. J. Pathol
.
139
,
787
799
.
Kumar
,
N. M.
,
Sigurdson
,
S. L.
,
Sheppard
,
D.
and
Lwebuga-Mukasa
,
J. S.
(
1995
).
Differential modulation of integrin receptors and extracellular matrix laminin by transforming growth factor-beta 1 in rat alveolar epithelial cells
.
Exp. Cell Res
.
221
,
385
394
.
Kurpakus
,
M. A.
,
Quaranta
,
V.
and
Jones
,
J. C.
(
1991
).
Surface relocation of alpha 6 beta 4 integrins and assembly of hemidesmosomes in an in vitro model of wound healing
.
J. Cell Biol
.
115
,
1737
1750
.
Lampe
,
P. D.
,
Nguyen
,
B. P.
,
Gil
,
S.
,
Usui
,
M.
,
Olerud
,
J.
,
Takada
,
Y.
and
Carter
,
W. G.
(
1998
).
Cellular interaction of integrin alpha3beta1 with laminin 5 promotes gap junctional communication
.
J. Cell Biol
.
143
,
1735
1747
.
Langhofer
,
M.
,
Hopkinson
,
S. B.
and
Jones
,
J. C.
(
1993
).
The matrix secreted by 804G cells contains laminin-related components that participate in hemidesmosome assembly in vitro
.
J. Cell Sci
.
105
,
753
764
.
Languino
,
L. R.
,
Gehlsen
,
K. R.
,
Wayner
,
E.
,
Carter
,
W. G.
,
Engvall
,
E.
and
Ruoslahti
.
E.
(
1989
).
Endothelial cells use alpha 2 beta 1 integrin as a laminin receptor
.
J. Cell Biol
.
109
,
2455
2462
.
Larjava
,
H.
,
Salo
,
T.
,
Haapasalmi
,
K.
,
Kramer
,
R. H.
and
Heino
,
J.
(
1993
).
Expression of integrins and basement membrane components by wound keratinocytes
.
J. Clin. Invest
.
92
,
1425
1435
.
Lichtner
,
R. B.
,
Howlett
,
A. R.
,
Lerch
,
M.
,
Xuan
,
J. A.
,
Brink
,
J.
,
Langton-Webster
,
B.
and
Schneider
M. R.
(
1998
).
Negative cooperativity between alpha3beta1 and alpha2beta1 integrins in human mammary carcinoma MDA MB 231 cells
.
Exp. Cell Res
.
240
,
368
376
.
Lochter
,
A.
,
Navre
,
M.
,
Werb
,
Z.
and
Bissell
,
M. J.
(
1999
).
Alpha1 and alpha2 integrins mediate invasive activity of mouse mammary carcinoma cells through regulation of stromelysin-1 expression
.
Mol. Biol. Cell
.
10
,
271
282
.
Marchisio
,
P. C.
,
Bondanza
,
S.
,
Cremona
,
O.
,
Cancedda
,
R.
and
De Luca
,
M.
(
1991
).
Polarized expression of integrin receptors (alpha 6 beta 4, alpha 2 beta 1, alpha 3 beta 1, and alpha v beta 5) and their relationship with the cytoskeleton and basement membrane matrix in cultured human keratinocytes
.
J. Cell Biol
.
112
,
761
773
.
Marinkovich
,
M. P.
,
Lunstrum
,
G. P.
and
Burgeson
,
R. E.
(
1992
).
The anchoring filament protein kalinin is synthesized and secreted as a high molecular weight precursor
.
J. Biol. Chem
.
267
,
17900
17906
.
Massague
,
J.
(
1990
).
The transforming growth factor-beta family
.
Annu. Rev. Cell Biol
.
6
,
597
641
.
Moses
,
H. L
,
Yang
,
E. Y.
and
Pietenpol
,
J. A.
(
1990
).
TGF-beta stimulation and inhibition of cell proliferation: new mechanistic insights
.
Cell
63
,
245
247
.
Miettinen
,
P. J.
,
Ebner
,
R.
,
Lopez
,
A. R.
and
Derynck
,
R.
(
1994
).
TGF-beta induced transdifferentiation of mammary epithelial cells to mesenchymal cells: involvement of type I receptors
.
J. Cell Biol
.
127
,
2021
2036
.
Mizushima
,
H.
,
Koshikawa
,
N.
,
Moriyama
,
K.
,
Takamura
,
H.
,
Nagashima
,
Y.
,
Hirahara
,
F.
and
Miyazaki
,
K.
(
1998
).
Wide distribution of laminin-5 gamma 2 chain in basement membranes of various human tissues
.
Horm. Res
.
2
,
7
14
.
Mizushima
,
H.
,
Takamura
,
H.
,
Miyagi
,
Y.
,
Kikkawa
,
Y.
,
Yamanaka
,
N.
,
Yasumitsu
,
H.
,
Misugi
,
K.
and
Miyazaki
,
K.
(
1997
).
Identification of integrin-dependent and -independent cell adhesion domains in COOH-terminal globular region of laminin-5 alpha 3 chain
.
Cell Growth Differ
.
8
,
979
987
.
Miyazaki
,
K.
,
Kikkawa
,
Y.
,
Nakamura
,
A.
,
Yasumitsu
,
H.
and
Umeda
,
M.
(
1993
).
A large cell-adhesive scatter factor secreted by human gastric carcinoma cells
.
Proc. Natl. Acad. Sci. USA
90
,
11767
11771
.
Nguyen
,
B. P.
,
Gil
,
S. G.
and
Carter
,
W. G.
(
2000
).
Deposition of laminin 5 by keratinocytes regulates integrin adhesion and signaling
.
J. Biol. Chem
.
275
,
31896
907
.
Nievers
,
M. G.
,
Schaapveld
,
R. Q.
and
Sonnenberg
,
A.
(
1999
).
Biology and function of hemidesmosomes
.
Matrix Biol
.
18
,
5
17
.
Nomizu
,
M.
,
Kuratomi
,
Y.
,
Song
,
S. Y.
,
Ponce
,
M. L.
,
Hoffman
,
M. P.
,
Powell
,
S. K.
,
Miyoshi
,
K.
,
Otaka
,
A.
,
Kleinman
,
H. K.
and
Yamada
,
Y.
(
1997
).
Identification of cell binding sequences in mouse laminin gamma1 chain by systematic peptide screening
.
J. Biol. Chem
.
272
,
32198
32205
.
Orian-Rousseau
,
V.
,
Aberdam
,
D.
,
Rousselle
,
P.
,
Messent
,
A.
,
Gavrilovic
,
J.
,
Meneguzzi
,
G.
,
Kedinger
,
M.
and
Simon-Assmann
,
P.
(
1998
).
Human colonic cancer cells synthesize and adhere to laminin-5. Their adhesion to laminin-5 involves multiple receptors among which is integrin alpha2beta1
.
J. Cell Sci
.
111
,
1993
2004
.
O’Toole
,
E. A.
,
Marinkovich
,
M. P.
,
Hoeffler
,
W. K.
,
Furthmayr
,
H.
and
Woodley
,
D. T.
(
1997
).
Laminin-5 inhibits human keratinocyte migration
.
Exp. Cell Res
.
233
,
330
339
.
Pfaff
,
M.
,
Gohring
,
W.
,
Brown
,
J. C.
and
Timpl
,
R.
(
1994
).
Binding of purified collagen receptors (alpha 1 beta 1, alpha 2 beta 1) and RGD-dependent integrins to laminins and laminin fragments
.
Eur. J. Biochem
.
225
,
975
984
.
Pilcher
,
B. K.
,
Dumin
,
J. A.
,
Sudbeck
,
B. D.
,
Krane
,
S. M.
,
Welgus
,
H. G.
and
Parks
,
W. C.
(
1997
).
The activity of collagenase-1 is required for keratinocyte migration on a type I collagen matrix
.
J. Cell Biol
.
137
,
1445
1457
.
Pyke
,
C.
,
Romer
,
J.
,
Kallunki
,
P.
,
Lund
,
L. R.
,
Ralfkiaer
,
E.
,
Dano
,
K.
and
Tryggvason
,
K.
(
1994
).
The gamma 2 chain of kalinin/laminin 5 is preferentially expressed in invading malignant cells in human cancers
.
Am. J. Pathol
.
145
,
782
791
.
Qin
,
P.
and
Kurpakus
,
M. A.
(
1998
).
The role of laminin-5 in TGF alpha/EGF-mediated corneal epithelial cell motility
.
Exp. Eye Res
.
66
,
569
579
.
Rousselle
,
P.
and
Aumailley
,
M.
(
1994
).
Kalinin is more efficient than laminin in promoting adhesion of primary keratinocytes and some other epithelial cells and has a different requirement for integrin receptors
.
J. Cell Biol
.
125
,
205
214
.
Rousselle
,
P.
,
Lunstrum
,
G. P.
,
Keene
,
D. R.
and
Burgeson
,
R. E.
(
1991
).
Kalinin: an epithelium-specific basement membrane adhesion molecule that is a component of anchoring filaments
.
J. Cell Biol
.
114
,
567
576
.
Ryan
,
M. C.
,
Lee
,
K.
,
Miyashita
,
Y.
and
Carter
,
W. G.
(
1999
).
Targeted disruption of the LAMA3 gene in mice reveals abnormalities in survival and late stage differentiation of epithelial cells
.
J. Cell Biol
.
145
,
1309
1323
.
Ryan
,
M. C.
,
Tizard
,
R.
,
VanDevanter
,
D. R.
and
Carter
,
W. G.
(
1994
).
Cloning of the LamA3 gene encoding the alpha 3 chain of the adhesive ligand epiligrin. Expression in wound repair
.
J. Biol. Chem
.
269
,
22779
22787
.
Salo
,
S.
,
Haakana
,
H.
,
Kontusaari
,
S.
,
Hujanen
,
E.
,
Kallunki
,
T.
and
Tryggvason
,
K.
(
1999
).
Laminin-5 promotes adhesion and migration of epithelial cells: identification of a migration-related element in the gamma2 chain gene (LAMC2) with activity in transgenic mice
.
Matrix Biol
.
18
,
197
210
.
Sheppard
,
D.
,
Cohen
,
D. S.
,
Wang
,
A.
and
Busk
,
M.
(
1992
).
Transforming growth factor beta differentially regulates expression of integrin subunits in guinea pig airway epithelial cells
.
J. Biol. Chem
.
267
,
17409
17414
.
Sonnenberg
,
A.
,
de Melker
,
A. A.
,
Martinez de Velasco
,
A. M.
,
Janssen
,
H.
,
Calafat
,
J.
and
Niessen
,
C. M.
(
1993
).
Formation of hemidesmosomes in cells of a transformed murine mammary tumor cell line and mechanisms involved in adherence of these cells to laminin and kalinin
.
J. Cell Sci
.
106
,
1083
1102
.
Sterk
,
L. M.
,
Geuijen
,
C. A
,
Oomen
,
L. C.
,
Calafat
,
J.
,
Janssen
,
H.
and
Sonnenberg
,
A.
(
2000
).
The tetraspan molecule CD151, a novel constituent of hemidesmosomes, associates with the integrin alpha6beta4 and may regulate the spatial organization of hemidesmosomes
.
J. Cell Biol
.
149
,
969
982
.
Symington
,
B. E.
,
Takada
,
Y.
and
Carter
,
W. G.
(
1993
).
Interaction of integrins alpha 3 beta 1 and alpha 2 beta 1: potential role in keratinocyte intercellular adhesion
.
J. Cell Biol
.
120
,
523
535
.
Tennenbaum
,
T.
,
Li
,
L.
,
Belanger
,
A. J.
,
De Luca
,
L. M.
and
Yuspa
,
S. H.
(
1996
).
Selective changes in laminin adhesion and alpha 6 beta 4 integrin regulation are associated with the initial steps in keratinocyte maturation
.
Cell Growth Differ
.
7
,
615
628
.
Vailly
,
J.
,
Verrando
,
P.
,
Champliaud
,
M. F.
,
Gerecke
,
D.
,
Wagman
,
D. W.
,
Baudoin
,
C.
,
Aberdam
,
D.
,
Burgeson
,
R.
,
Bauer
,
E.
and
Ortonne
,
J. P.
(
1994
).
The 100-kDa chain of nicein/kalinin is a laminin B2 chain variant
.
Eur. J. Biochem
.
219
,
209
218
.
Virolle
,
T.
,
Monthouel
,
M. N.
,
Djabari
,
Z.
,
Ortonne
,
J. P.
,
Meneguzzi
,
G.
and
Aberdam
,
D.
(
1998
).
Three activator protein-1-binding sites bound by the Fra-2.JunD complex cooperate for the regulation of murine laminin alpha3A (lama3A) promoter activity by transforming growth factor-beta
.
J. Biol. Chem
.
273
,
17318
17325
.
Weitzman
,
J. B.
,
Pasqualini
,
R.
,
Takada
,
Y.
and
Hemler
,
M. E.
(
1993
).
The function and distinctive regulation of VLA-3 in cell adhesion, spreading, and homotypic cell aggregation
.
J. Biol. Chem
.
268
,
8651
8657
.
Wu
,
C.
,
Chung
,
A. E.
and
McDonald
,
J. A.
(
1995
).
A novel role for alpha 3 beta 1 integrins in extracellular matrix assembly
.
J. Cell Sci
.
108
,
2511
2523
.
Zambruno
,
G.
,
Marchisio
,
P. C.
,
Marconi
,
A.
,
Vaschieri
,
C.
,
Melchiori
,
A.
,
Giannetti
,
A.
and
De Luca
,
M.
(
1995
).
Transforming growth factor-beta 1 modulates beta 1 and beta 5 integrin receptors and induces the de novo expression of the alpha v beta 6 heterodimer in normal human keratinocytes: implications for wound healing
.
J. Cell Biol
.
129
,
853
865
.
Zhang
,
K.
and
Kramer
,
R. H.
(
1996
).
Laminin 5 deposition promotes keratinocyte motility
.
Exp. Cell Res
.
227
,
309
322
.
Zicha
,
D.
,
Genot
,
E.
,
Dunn
,
G. A.
and
Kramer
I. M.
(
1999
).
TGFbeta1 induces a cell-cycle-dependent increase in motility of epithelial cells
.
J. Cell Sci
.
112
,
447
454
.