Src family kinase (SFK) is known to control various cell functions, but the significance of the location of its activation was largely unknown. We herein revealed that SFK activation occurs in lipid rafts. Based on this finding, we have developed a lipid raft–targeted SFK inhibitory fusion protein (LRT-SIFP) that inhibits the SFK activity in lipid rafts. LRT-SIFP has a peptide inhibitor of SFK and a lipid raft–targeting sequence in which two cysteine residues are palmitoylated for clustering in lipid rafts. LRT-SIFP was found to inhibit cell adhesion and cell cycle progression of human breast cancer cell lines MCF-7 and MDA-MB231. On the other hand, the cell functions of MCF-7 cells were found to be not affected with a previously developed peptide inhibitor of SFK that lacks the lipid raft–targeting sequence. In addition, when we replaced the targeting sequence of LRT-SIFP with the consensus sequence for geranylgeranylation to make LRT-SIFP unable to cluster in lipid rafts, this mutated LRT-SIFP did not show any effect on the above cell functions of MCF-7 cells. Furthermore, in contrast to the breast cancer cell lines, LRT-SIFP did not show any inhibitory effect on cell adhesion and cell cycle progression of human normal cell line HEK293. The present lipid raft–specific knockdown of SFK activity would potentially be useful for selective cancer therapy to prevent tumorigenesis and metastasis of breast cancer cells. [Cancer Res 2007;67(17):8139–48]

Cholesterol-enriched nanodomains called lipid rafts are thought to act as a platform for protein signaling in cells, but the physiologic significance of lipid rafts in cells and tissues has remained unsolved (1, 2). The main point of the present work is to show the physiologic significance of kinase activity in lipid rafts. Src family kinases (SFK) are known to be ubiquitously distributed in the cell membranes and regulate many biological processes (3, 4). To locate with high resolution where the SFK activation occurs in the cell membranes, we developed a genetically encoded transmembrane fluorescent indicator for detecting the SFK activation at the cell membranes and named it TM-Srcus (Fig. 1A; refs. 5, 6). TM-Srcus can monitor the substrate phosphorylation by activated SFK as the decrease in the cyan fluorescent protein (CFP)/yellow fluorescent protein (YFP) emission ratio through fluorescence resonance energy transfer (FRET; Fig. 1A). The detailed principle of this indicator is described in Materials and Methods. The total internal reflection fluorescence imaging of the SFK activation on the plasma membrane by TM-Srcus showed that SFK activation takes place in lipid rafts.

Figure 1.

TM-Srcus, a fluorescent indicator for detecting SFK activation in cell membranes. A, left, the principle of TM-Srcus for visualizing SFK activation in cell membranes. On SFK activation, a conformational change in TM-Srcus occurs due to binding of a tyrosine phosphorylation recognition (SH2) domain to a phosphorylated SFK substrate (Y314) domain, which results in an intramolecular FRET response. A, right, constructed cDNAs of TM-Srcus and TM-SrcusY314A. TM-Srcus is a tandem fusion protein that consists of seven parts: a transmembrane domain, a CFP, the Y314 domain containing the tyrosine phosphorylation site (red-colored Y), a flexible linker sequence (Ln), the SH2 domain, a YFP, and a NES. The Y314A domain is a mutated SFK substrate domain containing the alanine mutation site (red-colored A). B, time courses of the CFP/YFP emission ratio of TM-Srcus in single MCF-7 cells observed under a conventional fluorescence microscope. The stimulation of cells with 10 nmol/L E2 provokes a rapid and significant decrease in the CFP/YFP emission ratio (filled square). After pretreatment with a specific SFK inhibitor of 5 nmol/L PP2 for 1 h, no significant change in the CFP/YFP emission ratio was observed on 10 nmol/L E2 stimulation (green filled triangle). Pretreatment of cells with 1 μmol/L ICI 182,789 or transfection of cells with the selective siRNA against Src inhibited the E2-induced decrease in the CFP/YFP emission ratio (red open diamond and blue open circle). The region enclosed by a white-colored line represents the single MCF-7 cell. Representative results of three independent measurements. C, relative decreases in CFP/YFP emission ratio of TM-Srcus in single MCF-7 cells [columns, mean (n = 3); bars, SD]. Inset, anti-Src immunoblot analysis of cell lysate from MCF-7 cells transfected with or without siRNA against Src. Taken together, the results in (B) and (C) indicate that the fluorescence responses of TM-Srcus accurately represent the substrate phosphorylation by activated SFK on stimulation with E2. D, pseudocolor images of the CFP/YFP emission ratio of TM-Srcus in the plasma membrane of the MCF-7 cell under a TIRFM. The images were captured before and 20 min after the stimulation of the cell with 10 nmol/L E2. The region enclosed by a white line represents the plasma membrane of the single MCF-7 cell. Representative results of three independent measurements. B to D, MCF-7 cells expressing TM-Srcus were made quiescent by incubation for 12 h with medium supplemented with DCC-treated serum before E2 stimulation.

Figure 1.

TM-Srcus, a fluorescent indicator for detecting SFK activation in cell membranes. A, left, the principle of TM-Srcus for visualizing SFK activation in cell membranes. On SFK activation, a conformational change in TM-Srcus occurs due to binding of a tyrosine phosphorylation recognition (SH2) domain to a phosphorylated SFK substrate (Y314) domain, which results in an intramolecular FRET response. A, right, constructed cDNAs of TM-Srcus and TM-SrcusY314A. TM-Srcus is a tandem fusion protein that consists of seven parts: a transmembrane domain, a CFP, the Y314 domain containing the tyrosine phosphorylation site (red-colored Y), a flexible linker sequence (Ln), the SH2 domain, a YFP, and a NES. The Y314A domain is a mutated SFK substrate domain containing the alanine mutation site (red-colored A). B, time courses of the CFP/YFP emission ratio of TM-Srcus in single MCF-7 cells observed under a conventional fluorescence microscope. The stimulation of cells with 10 nmol/L E2 provokes a rapid and significant decrease in the CFP/YFP emission ratio (filled square). After pretreatment with a specific SFK inhibitor of 5 nmol/L PP2 for 1 h, no significant change in the CFP/YFP emission ratio was observed on 10 nmol/L E2 stimulation (green filled triangle). Pretreatment of cells with 1 μmol/L ICI 182,789 or transfection of cells with the selective siRNA against Src inhibited the E2-induced decrease in the CFP/YFP emission ratio (red open diamond and blue open circle). The region enclosed by a white-colored line represents the single MCF-7 cell. Representative results of three independent measurements. C, relative decreases in CFP/YFP emission ratio of TM-Srcus in single MCF-7 cells [columns, mean (n = 3); bars, SD]. Inset, anti-Src immunoblot analysis of cell lysate from MCF-7 cells transfected with or without siRNA against Src. Taken together, the results in (B) and (C) indicate that the fluorescence responses of TM-Srcus accurately represent the substrate phosphorylation by activated SFK on stimulation with E2. D, pseudocolor images of the CFP/YFP emission ratio of TM-Srcus in the plasma membrane of the MCF-7 cell under a TIRFM. The images were captured before and 20 min after the stimulation of the cell with 10 nmol/L E2. The region enclosed by a white line represents the plasma membrane of the single MCF-7 cell. Representative results of three independent measurements. B to D, MCF-7 cells expressing TM-Srcus were made quiescent by incubation for 12 h with medium supplemented with DCC-treated serum before E2 stimulation.

Close modal

Based on this finding, we herein extended to develop a lipid raft–targeted SFK inhibitory fusion protein (LRT-SIFP) to inhibit SFK activity in lipid rafts. The LRT-SIFP contains the peptide inhibitor of SFK and the targeting sequence for localizing the SIFP to lipid rafts. The significance about the subcellular locations of kinase activity has hardly been studied by conventional inhibition methods such as small interfering RNA (siRNA) and chemical or peptide inhibitors, which ubiquitously inhibit kinase activity in cells. The present LRT-SIFP elucidated the importance of the SFK activation in lipid rafts for function of breast cancer cells.

The previously developed peptide inhibitor of SFK does not affect cell functions of MCF-7 cells derived from human breast cancer, although it has high potency. In contrast to this conventional peptide inhibitor, the present LRT-SIFP inhibits cell adhesion and cell cycle progression of MCF-7 and MDA-MB231 cells. In addition, these inhibitory effects of LRT-SIFP on cell functions are specific for tumor cell lines. The lipid raft–specific knockdown of SFK activity would potentially be useful for selective cancer therapy to prevent tumorigenesis and metastasis of breast cancer cells.

The principle of TM-Srcus. TM-Srcus is a fusion protein that contains a SFK substrate domain, a phosphorylation recognition domain, a flexible linker sequence, and two green fluorescent protein (GFP) mutants. When phosphorylated at the tyrosine in the SFK substrate domain of TM-Srcus by activated SFK, the adjacent phosphorylation recognition domain specifically binds to this phosphorylated tyrosine. When this binding occurs, the distance between CFP and YFP in TM-Srcus is expected to become shorter, and an intramolecular FRET response results (Fig. 1A). The emission ratio is the CFP fluorescence intensity divided by the YFP fluorescence intensity. If FRET is induced, the CFP fluorescence intensity decreases and the YFP fluorescence intensity increases. The emission ratio is decreased as a result. The indicator thereby monitors the SFK activation in vivo as the decrease in the CFP/YFP emission ratio through FRET. To directly detect the SFK activation in the whole region of cell membranes, TM-Srcus has the transmembrane domain that possesses a high affinity for cell membranes. The indicator can thus monitor SFK activation throughout the cytoplasmic surfaces of the cell membranes (Fig. 1A).

Plasmid construction. To construct the genes of TM-Srcus and TM-SrcusY314A, cDNA fragments of a transmembrane domain from a transmembrane phosphoprotein (Cbp, amino acids 1–52; ref. 7), the Y314 domain from a phosphopeptide containing Tyr314 of Cbp fused with Ln, the Y314A domain fused with Ln, the SH2 domain from a COOH-terminal Src kinase (Csk, amino acids 80–162; ref. 8), CFP, and YFP with nuclear export signal sequence (NES) from a HIV-derived protein (9) were generated by PCR and cloned into pBlueScript SK(+).

To construct the genes of LRT-SIFP and its alanine and deletion mutants, cDNA fragments of the lipid raft–targeting sequence (COOH-terminal H-Ras, CMSCKCVLS) fused with Ln, the mutated lipid raft–targeting sequence (GGGS with COOH-terminal Rho-A, GCLVL) fused with Ln, the peptide inhibitor for SFK (MIYKYYF) fused with a flag tag, the alanine-mutated peptide inhibitor for SFK (MIYKYAF) fused with a flag tag, and YFP were generated by PCR and cloned into pBlueScript SK(+).

All cloning enzymes (Takara Biomedical) were used according to the manufacturer's instructions. The PCR fragments were sequenced with an ABI310 genetic analyzer (Applied Biosystems). The constructs were subcloned into pcDNA3.1(+) (Invitrogen) behind a Kozak sequence using HindIII and XhoI sites.

Imaging of cells. MCF-7 cells expressing TM-Srcus were starved with a steroid-free medium [phenol red–free Eagle's MEM with 2% dextran-coated charcoal (DCC)–treated serum] for 12 h, then washed twice with HBSS (Sigma). For a total internal reflection fluorescence imaging, cells were observed under a total internal reflection fluorescence microscope (TIRFM; IX70, Olympus) with a charge-coupled device camera CoolSnap ES (Roper Scientific) controlled by MetaFluor (Universal Imaging). The exposure time at 440 ± 10-nm excitation was 300 ms. The fluorescent images of CFP and YFP were obtained with filters at 480 ± 15 and 535 ± 12.5 nm with a 60× oil immersion objective PlanApo60 (Olympus). For a conventional fluorescence imaging, cells were pretreated with PP2 (Calbiochem), stimulated with 17β-estradiol (E2; Sigma), and imaged at room temperature under a Carl Zeiss Axiovert 135 microscope (Carl Zeiss) with a cooled charge-coupled device camera MicroMax (Roper Scientific) controlled by MetaFluor. The exposure time at 440 ± 10-nm excitation was 200 ms. The fluorescent images were obtained with filters at 480 ± 15 and 535 ± 12.5 nm with a 40× oil immersion objective (Carl Zeiss).

Density gradient fractionation. MCF-7 cells in a 10-cm dish expressing TM-Srcus and LRT-SIFP were scraped in ice-cold HBSS and spun down at 2,000 rpm 4°C, lysed thoroughly by pipetting through a 200-μL yellow pipetting tip with 180 μL TNE [10 mmol/L Tris-HCl (pH 7.6), 500 mmol/L NaCl, 1 mmol/L EDTA], 1% Triton X-100, 10% sucrose, 2 mmol/L sodium orthovanadate, 1% protease inhibitor cocktail (Sigma) at 4°C, and incubated for 20 min on ice. Three hundred sixty microliters of cold 60% Optiprep (Axis-Shield PoC AS) were added to the extract and incubated for 10 min on ice. The mixture of the extract and 60% Optiprep was transferred into thick-walled polycarbonate tubes (Beckman Coulter). The sample was overlaid with a 540-μL step of each of 35%, 30%, 25%, 20%, 0% Optiprep in TNE, 1% Triton X-100, 10% sucrose, 2 mmol/L sodium orthovanadate. The gradients were spun for 4 h at 200,000 × g at 4°C and collected into six 540-μL fractions.

Immunoprecipitation and immunoblot analysis. For immunoprecipitation, the six gradient fractions were diluted with an equal volume of TNE, 1% Triton X-100, 10% sucrose, and 2 mmol/L sodium orthovanadate (10). The diluted samples were immunoprecipitated with anti-phosphotyrosine antibody (PY20, Santa Cruz Biotechnology). For immunoblotting, the samples were separated by electrophoresis on a 10% SDS-acrylamide gel and the electrophoresed proteins were transferred onto a nitrocellulose membrane. The membranes were blocked with 1% dry milk or 3% bovine serum albumin (BSA) in TBST and was incubated with primary antibodies: anti-phosphotyrosine (PY20), anti-Fyn (Fyn 3, Santa Cruz Biotechnology), anti–transferrin receptor (Zymed Laboratories), anti-GFP (Clontech), anti–caveolin-1 (BD Transduction Laboratories), anti-Yes (1B7, WAKO Biochemicals), and anti-Src (GD11, Upstate Biotechnology). The anti–active Src antibody (clone 28) was kindly provided by Dr. K. Owada (Kyoto Pharmaceutical University, Yamashinaku, Kyoto, Japan). Bands were visualized with horseradish peroxidase (HRP)–conjugated antirabbit or antimouse immunoglobulin G (Amersham Life Science).

Cell culture and transfection. MCF-7 cells were cultured in MEM (Sigma) supplemented with 10% fetal bovine serum (FBS), 1% penicillin/streptomycin, and 0.1 mmol/L nonessential amino acids at 37°C in 5% CO2. MDA-MB231 cells were cultured in DMEM supplemented with 10% FBS and 2 mmol/L l-glutamine at 37°C in 5% CO2. HEK293 cells were cultured in DMEM supplemented with 10% FBS at 37°C in 5% CO2. The cells were transfected with LipofectoAMINE 2000 (Invitrogen). Cells were plated 24 to 36 h before transfection onto glass-bottom dishes for fluorescence imaging or onto plastic culture dishes for immunoblot analysis.

Cell staining with cholera toxin B subunit-Alexa647. MCF-7 cells expressing TM-Srcus and LRT-SIFP were washed with HBSS and incubated at 37°C for 1 h with 2 μg/mL cholera toxin B subunit (CTXB)-Alexa647 (Molecular Probes) in MEM supplemented with 0.01% BSA, 1% penicillin/streptomycin, 25 mmol/L HEPES, and 0.1 mmol/L nonessential amino acids (11). After staining, cells were washed twice with HBSS and observed at room temperature under a TIRFM or a confocal microscope. The exposure time at 633 ± 10-nm excitation was 200 ms. The fluorescent images of CTXB-Alexa647 under a TIRFM or a confocal microscope were obtained with a 60× oil immersion objective PlanApo 60 and a 100× oil immersion objective, respectively.

Cell adhesion assay. MCF-7 cells expressing LRT-SIFP were washed with PBS, trypsinized, and resuspended with PBS. Aliquots of the cell suspension (106 per mL) were placed on a 24 × 24-mm micro-cover glass (Matsunami) and on the bottom of a six-well plate (Nunc). After 24-h incubation, 80% confluent cells both on the cover slide and on the bottom of the well were transfected with each of constructed cDNAs encoding the SIFPs and were incubated for 24 h at 37°C. For counting the number of originally plated fluorescence-positive cells, the transfected cells on the cover slide were washed with HBSS and fixed with 3% PFA for 10 min at 4°C. The fluorescence-positive cells on the cover slide were directly counted under a fluorescence microscope (×40). For counting the number of adhered fluorescence-positive cells, the transfected cells on the bottom of the well were trypsinized and resuspended in culture medium. The whole-cell suspension (106 cells/mL) was placed on the cover slide coated with 33 μg/mL fibronectin in PBS overnight at 4°C and was incubated for 4 h at 37°C with medium containing 10% FBS (12). The adhered cells on the fibronectin-coated cover slide were washed with HBSS and fixed with 3% PFA for 10 min at 4°C. The adhered fluorescence-positive cells were directly counted under a fluorescence microscope (×40). The adhesion index is the number of the adhered fluorescence-positive cells divided by that of the originally plated fluorescence-positive cells. The adhesion index of MCF-7 cells with and without incubation with 5 μmol/L PP2 for 4 h at 37°C (Supplementary Fig. S1A) was measured under a microscope (×40) as described above.

Propidium iodide staining and fluorescence-activated cell sorting analysis. MCF-7 cells (1.5 × 106) expressing LRT-SIFP were washed with PBS, trypsinized, and resuspended with ice-cold PBS. The resuspended cells were fixed and permeabilized with 70% ethanol for 30 min at 4°C. The permeabilized cells were washed twice with PBS and incubated with 100 μg/mL RNase A (Qiagen) in PBS for 30 min at room temperature, and then subjected to propidium iodide staining (50 μg/mL) for 30 min at 4°C. The cell cycle profile of MCF-7 cells with and without incubation with 5 μmol/L PP2 (Supplementary Fig. S2) was obtained as described above.

MCF-7 cells (1.5 × 106) expressing SIFP or YFP were fixed with 1% PFA in PBS for 15 min at 4°C to avoid a drain of the expressing protein from the cells through permeabilization. The PFA-fixed cells were then permeabilized and stained with propidium iodide using 500 μL of low-salt solution (0.1% Triton X-100, 3% polyethylene glycol-8000, 50 μg/mL propidium iodide, 100 μg/mL RNase, 4 mmol/L sodium citrate, pH 7.1–7.2) for 30 min at 4°C, and subsequently added with an equal volume of high-salt solution (0.1% Triton X-100, 3% polyethylene glycol-8000, 50 μg/mL propidium iodide, 100 μg/mL RNase, 400 mmol/L NaCl, pH 7.1–7.2) for 30 min at 4°C (13). The propidium iodide–stained cells were analyzed by fluorescence-activated cell sorting (Beckman Coulter).

TM-Srcus can monitor SFK activation induced by a physiologic dose of 10 nmol/L estrogen in living MCF-7 cells. We constructed cDNA that encodes TM-Srcus (Fig. 1A,, right) and introduced the cDNA in MCF-7 cells, which are derived from human female breast cancer cells. TM-Srcus was observed in the plasma membrane and endomembranes (i.e., ER and Golgi membranes) owing to its transmembrane domain (Fig. 1A and B). We stimulated the cells expressing TM-Srcus with a physiologic dose of 10 nmol/L E2, which is known to activate SFK through estrogen receptor outside the nucleus (14). The CFP/YFP emission ratio of TM-Srcus decreased on 10 nmol/L E2 stimulation, and this decrease in the emission ratio reached a plateau within 20 min (Fig. 1B). After pretreatment of the cells with a specific SFK inhibitor, 5 nmol/L PP2 for 1 h, no significant change in the CFP/YFP emission ratio was observed even on 10 nmol/L E2 stimulation (Fig. 1B and C). In addition, 10 nmol/L E2 did not induce a change in the CFP/YFP emission ratio in the cells cotransfected with TM-Srcus and selective siRNA against Src (Fig. 1B and C). In addition, pretreatment of the cells with antiestrogenic agent ICI 182,780 for 1 h inhibited the E2-induced change in the CFP/YFP emission ratio of TM-Srcus (Fig. 1B and C). TM-SrcusY314A in which the tyrosine was replaced with alanine that has no phosphoacceptor site (Fig. 1A,, right) did not show any change in the CFP/YFP emission ratio on 10 nmol/L E2 stimulation (Fig. 1C). These results indicate that TM-Srcus can monitor substrate phosphorylation by activated SFK in MCF-7 cells stimulated with a physiologic dose of 10 nmol/L E2.

Lipid raft–specific SFK activation in MCF-7 cells revealed by TM-Srcus. We next observed MCF-7 cells expressing TM-Srcus under a TIRFM. TIRFM provides a fluorescent image within a depth of ∼100 nm from the contact regions, enabling a selective visualization of the basal plasma membrane of cells (15). Pseudocolor images that represent the CFP/YFP emission ratio of TM-Srcus showed a blue shift in specific regions of the plasma membrane on 10 nmol/L E2 stimulation (Fig. 1D). These spatially limited regions of the plasma membrane may be clarified by a course of experiments described in the following. A single lipid raft cannot be observed because of its nanometer size by conventional fluorescence microscopy with wavelength limitation of 200 nm, but a lipid raft cluster consisting of multiple rafts was what we observed. We stained MCF-7 cells with a lipid raft marker, CTXB-Alexa647 (1), and observed the stained cells under a TIRFM. The E2-induced SFK activation occurred locally in the plasma membranes as was observed in the cells without staining by CTXB-Alexa647 (Figs. 1D and 2,A, left and middle). The blue-shifted regions that represent the SFK activation seem to be located where CTXB is concentrated (Fig. 2,A, middle and right). A scatterplot of the emission ratio of TM-Srcus versus the CTXB fluorescence intensity showed that a decrease in the emission ratio of TM-Srcus is correlated with an increase in the CTXB fluorescence intensity (R = −0.82; Fig. 2B). These results indicate that the activation of SFK on the stimulation of MCF-7 cells with E2 is provoked in lipid raft clusters in the plasma membrane.

Figure 2.

SFK activation in lipid rafts in MCF-7 cells. A, pseudocolor images of the CFP/YFP emission ratio of TM-Srcus in the plasma membrane of the MCF-7 cell stained with CTXB-Alexa647 under a TIRFM. The region enclosed by a white line represents the plasma membrane of the single MCF-7 cell. B, a scatterplot obtained from 70 regions of interest in the basal plasma membrane of the E2-stimulated cell expressed with TM-Srcus and stained with CTXB-Alexa647. The correlation between a decrease in emission ratio of TM-Srcus and an increase in CTXB fluorescence intensity was observed (R = −0.82). C, Western blot analysis of six density gradient fractions from TM-Srcus (68 kDa)–expressing MCF-7 cells stimulated with 10 nmol/L E2, 1 μmol/L P4, and 1 μmol/L dihydrotestosterone (DHT). The fractions were blotted with HRP-conjugated CTXB or antibodies against caveolin-1, transferrin receptor (TfR), GFP, and phosphotyrosine. D, Western blot analysis of six density gradient fractions from MCF-7 cells stimulated with 10 nmol/L E2. The fractions were blotted with HRP-conjugated CTXB or antibodies against caveolin-1, transferrin receptor, active Src, Src, Yes, and Fyn. A to D, MCF-7 cells were made quiescent by incubation for 12 h with medium supplemented with DCC-treated serum before steroid stimulation. Representative results of three independent measurements.

Figure 2.

SFK activation in lipid rafts in MCF-7 cells. A, pseudocolor images of the CFP/YFP emission ratio of TM-Srcus in the plasma membrane of the MCF-7 cell stained with CTXB-Alexa647 under a TIRFM. The region enclosed by a white line represents the plasma membrane of the single MCF-7 cell. B, a scatterplot obtained from 70 regions of interest in the basal plasma membrane of the E2-stimulated cell expressed with TM-Srcus and stained with CTXB-Alexa647. The correlation between a decrease in emission ratio of TM-Srcus and an increase in CTXB fluorescence intensity was observed (R = −0.82). C, Western blot analysis of six density gradient fractions from TM-Srcus (68 kDa)–expressing MCF-7 cells stimulated with 10 nmol/L E2, 1 μmol/L P4, and 1 μmol/L dihydrotestosterone (DHT). The fractions were blotted with HRP-conjugated CTXB or antibodies against caveolin-1, transferrin receptor (TfR), GFP, and phosphotyrosine. D, Western blot analysis of six density gradient fractions from MCF-7 cells stimulated with 10 nmol/L E2. The fractions were blotted with HRP-conjugated CTXB or antibodies against caveolin-1, transferrin receptor, active Src, Src, Yes, and Fyn. A to D, MCF-7 cells were made quiescent by incubation for 12 h with medium supplemented with DCC-treated serum before steroid stimulation. Representative results of three independent measurements.

Close modal

We next carried out a density gradient fractionation that separates the total lysates of the cells expressing TM-Srcus into six gradient fractions (see Materials and Methods; refs. 16, 17). This fractionation method enables the isolation of detergent-resistant membranes (DRM) that are not equated to lipid rafts but include part of a raft-associated protein (18). The gradient fractions were blotted with HRP-conjugated CTXB and anti–caveolin-1 antibody, which are known to bind to positive markers for DRMs, GM1 and caveolin, respectively (19). These gradient fractions were also blotted with antibody against transferrin receptor, a negative marker for DRMs (20). The distribution of the positive and negative markers for DRMs in these fractions was similar to what was previously observed in another epithelial cell line (ref. 20; Fig. 2C and D). We identified fractions 2, 3, and 4 as DRM fractions, and fraction 5 as a non-DRM fraction (Fig. 2C and D). From the immunoblotting of the six fractions with anti-GFP antibody, TM-Srcus was found to distribute in all fractions (Fig. 2C). However, the immunoblotting of these six fractions with anti-phosphotyrosine antibody showed that phosphorylation of TM-Srcus by activated SFK on 10 nmol/L E2 stimulation is induced only in fraction 4 (Fig. 2C). Furthermore, we carried out immunoblotting of these fractions with anti–active Src and anti-Src antibodies. The anti–active Src antibody is known to selectively recognize the COOH-terminal regulatory domain of the active Src (21). On 10 nmol/L E2 stimulation, activated Src was localized in the DRM fraction (fraction 4) in which TM-Srcus is phosphorylated (Fig. 2C and D), although Src was distributed in both DRM and non-DRM fractions (Fig. 2D). These results support the above conclusion that E2 provokes activation of SFK exclusively in lipid rafts in MCF-7 cells. In addition, this lipid raft–specific phosphorylation of TM-Srcus was elicited with other sex steroids, such as progesterone (P4) and androgen 5α-dihydrotestosterone (Fig. 2C), which are known to activate SFK through their receptors (14). It is thus concluded that the sex steroids, E2, P4, and dihydrotestosterone, provoke activation of SFK only in lipid rafts in MCF-7 cells.

We also examined whether the distribution of SFKs such as Src, Yes, and Fyn in the gradient fractions is changed by E2 stimulation. We carried out the immunoblotting of the gradient fractions with anti-Src, anti-Yes, and anti-Fyn antibodies. The distribution of Yes and Fyn in the gradient fractions was not changed on stimulation of MCF-7 cells with 10 nmol/L E2, but Src was found to translocate into DRM fraction on E2 stimulation (Fig. 2D). These results imply not only that Src in DRM fraction is activated by E2 but also that other Src located in non-DRM fraction is recruited to DRMs on E2 stimulation for its activation there.

The detailed structure of the LRT-SIFP. SFK activation has been known to be necessary for cell functions such as cell adhesion to integrin ligands and growth factor–induced mitogenesis (22, 23), which are closely related to growth and metastasis of cancer cells (24) and to homeostasis of normal cells. To determine whether the lipid raft–specific SFK activation has significant effects on these cell functions, we developed a LRT-SIFP that inhibits the SFK activity in lipid rafts. The LRT-SIFP consists of a pseudosubstrate-based peptide inhibitor of SFK (MIYKYYF; ref. 25), YFP, a flag tag, and a lipid raft–targeting sequence (Fig. 3A). The lipid raft–targeting sequence is the COOH-terminal nine amino acids of H-Ras (CMSCKCVLS) to which GFP was fused for visualizing lipid rafts in cells in previous studies (2628). This targeting sequence is known to be posttranslationally double-palmitoylated at the cysteine residues to localize in lipid rafts in the cells (26, 27).

Figure 3.

LRT-SIFP. A, constructed cDNAs of LRT-SIFP, LRT-SIFP Y6A, LRT-SIFP Del as LRT-YFP control, SIFP, SIFP Del as YFP control, mLRT-SIFP, and mLRT-SIFP Del as mLRT-YFP control. The construct of TM-Srcus and LRT-SIFP devoid of YFP connected by IRES sequence and the construct of TM-Srcus and mLRT-SIFP devoid of YFP connected by IRES sequence are also shown. LRT-SIFP consists of YFP, a flag tag, a peptide inhibitor for SFK containing the tyrosine phosphorylation site (red-colored Y), and the lipid raft–targeting sequence. LRT-SIFP Y6A contains the alanine-mutated peptide inhibitor containing the alanine mutation site (blue-colored A). LRT-SIFP Del (=LRT-YFP) has the peptide inhibitor for SFK deleted. SIFP is the YFP-fused peptide inhibitor of SFK that lacks the lipid raft–targeting sequence. SIFP Del is a YFP control. mLRT-SIFP has the mutated lipid raft–targeting sequence to reduce its raft association. mLRT-SIFP Del is a mLRT-YFP control. B, top, fluorescent images of LRT-SIFP and CTXB-Alexa647 in MCF-7 cells observed under a confocal fluorescence microscope. The YFP image in LRT-SIFP is shown in green. Staining with CTXB-Alexa647 is in red. The merged image is also shown. Bottom, zoomed images from the white rectangles on top. LRT-SIFP is colocalized with CTXB-Alexa647 in the plasma membrane. C, relative decrease in emission ratio of TM-Srcus on E2 stimulation in cells expressing TM-Srcus alone and in cells coexpressing TM-Srcus and LRT-SIFP [columns, mean (n = 3); bars, SD]. MCF-7 cells were made quiescent by incubation for 12 h with medium supplemented with DCC-treated serum. D, Western blot analysis of six density gradient fractions from MCF-7 cells expressing LRT-SIFP (34 kDa). The cell lysates of MCF-7 cells expressing LRT-SIFP were subjected to gradient fractionation, and the six density gradient fractions were obtained. The fractions were blotted with HRP-conjugated CTXB (positive marker for DRMs) and anti–transferrin receptor antibody (negative marker for DRMs). The six fractions were immunoprecipitated with anti-phosphotyrosine (pTyr) antibody and immunoblotted with anti-GFP antibody (see Materials and Methods). Treatment with the SFK-specific inhibitor PP2 (5 μmol/L) was done for 2 h at 37°C. Anti–active Src and anti-Src immunoblot analyses of the gradient fractions obtained from MCF-7 cells transfected with or without LRT-SIFP were also shown. B and D, MCF-7 cells transfected with LRT-SIFP were incubated for 24 h with medium containing 10% FBS before sample collection; representative results of three independent measurements.

Figure 3.

LRT-SIFP. A, constructed cDNAs of LRT-SIFP, LRT-SIFP Y6A, LRT-SIFP Del as LRT-YFP control, SIFP, SIFP Del as YFP control, mLRT-SIFP, and mLRT-SIFP Del as mLRT-YFP control. The construct of TM-Srcus and LRT-SIFP devoid of YFP connected by IRES sequence and the construct of TM-Srcus and mLRT-SIFP devoid of YFP connected by IRES sequence are also shown. LRT-SIFP consists of YFP, a flag tag, a peptide inhibitor for SFK containing the tyrosine phosphorylation site (red-colored Y), and the lipid raft–targeting sequence. LRT-SIFP Y6A contains the alanine-mutated peptide inhibitor containing the alanine mutation site (blue-colored A). LRT-SIFP Del (=LRT-YFP) has the peptide inhibitor for SFK deleted. SIFP is the YFP-fused peptide inhibitor of SFK that lacks the lipid raft–targeting sequence. SIFP Del is a YFP control. mLRT-SIFP has the mutated lipid raft–targeting sequence to reduce its raft association. mLRT-SIFP Del is a mLRT-YFP control. B, top, fluorescent images of LRT-SIFP and CTXB-Alexa647 in MCF-7 cells observed under a confocal fluorescence microscope. The YFP image in LRT-SIFP is shown in green. Staining with CTXB-Alexa647 is in red. The merged image is also shown. Bottom, zoomed images from the white rectangles on top. LRT-SIFP is colocalized with CTXB-Alexa647 in the plasma membrane. C, relative decrease in emission ratio of TM-Srcus on E2 stimulation in cells expressing TM-Srcus alone and in cells coexpressing TM-Srcus and LRT-SIFP [columns, mean (n = 3); bars, SD]. MCF-7 cells were made quiescent by incubation for 12 h with medium supplemented with DCC-treated serum. D, Western blot analysis of six density gradient fractions from MCF-7 cells expressing LRT-SIFP (34 kDa). The cell lysates of MCF-7 cells expressing LRT-SIFP were subjected to gradient fractionation, and the six density gradient fractions were obtained. The fractions were blotted with HRP-conjugated CTXB (positive marker for DRMs) and anti–transferrin receptor antibody (negative marker for DRMs). The six fractions were immunoprecipitated with anti-phosphotyrosine (pTyr) antibody and immunoblotted with anti-GFP antibody (see Materials and Methods). Treatment with the SFK-specific inhibitor PP2 (5 μmol/L) was done for 2 h at 37°C. Anti–active Src and anti-Src immunoblot analyses of the gradient fractions obtained from MCF-7 cells transfected with or without LRT-SIFP were also shown. B and D, MCF-7 cells transfected with LRT-SIFP were incubated for 24 h with medium containing 10% FBS before sample collection; representative results of three independent measurements.

Close modal

Characterization of LRT-SIFP. MCF-7 cells expressing LRT-SIFP were stained with the lipid raft marker CTXB-Alexa647 and were observed under a confocal microscope. Lipid raft clusters were observed in endosome-like vesicles and in the plasma membrane (Fig. 3B). LRT-SIFP was colocalized with the lipid raft clusters in the plasma membrane (Fig. 3B). We next analyzed the inhibitory effect of LRT-SIFP on SFK activity in lipid rafts by measuring the FRET response of TM-Srcus in the cells coexpressed with both TM-Srcus and LRT-SIFP. The cDNAs of TM-Srcus and LRT-SIFP lacking YFP were connected by the internal ribosome entry site (IRES) sequence and were transfected into MCF-7 cells (Fig. 3A). The E2-induced FRET response of TM-Srcus that represents the SFK activation in lipid rafts was completely inhibited by LRT-SIFP (Fig. 3C). This result indicates the inhibitory effect of LRT-SIFP on SFK activity in lipid rafts. This complete inhibition of the E2-induced SFK activation in lipid rafts by LRT-SIFP and the colocalization of LRT-SIFP with lipid raft clusters in the plasma membrane confirm that the FRET response of TM-Srcus observed with TIRFM (Fig. 2A) is due to the E2-induced SFK activation in lipid rafts in the plasma membrane, not in the endosome-like vesicles.

In addition, to examine the effect of palmitoylation of LRT-SIFP on SFK activity in lipid rafts, we replaced the targeting sequence of LRT-SIFP with the consensus sequence for geranylgeranylation (CLVL). The GFP fused with this consensus sequence does not cluster in lipid rafts (26). We named this mutated construct mLRT-SIFP (Fig. 3A). The IRES-connected cDNAs of TM-Srcus and mLRT-SIFP devoid of YFP (Fig. 3A) were transfected into MCF-7 cells. The cells coexpressing TM-Srcus and mLRT-SIFP were observed under a conventional epifluorescence microscope. mLRT-SIFP did not inhibit the E2-induced FRET response of TM-Srcus, which is due to the lipid raft–specific SFK activation (Fig. 3C). This result indicates that the palmitoylation of peptide inhibitor of SFK is necessary for the effective inhibition of the SFK activity in lipid rafts.

The inhibitory effect of LRT-SIFP on SFK activity in lipid rafts was also assessed by the density gradient fractionation method. LRT-SIFP contains the pseudosubstrate-based peptide inhibitor that is preferentially phosphorylated by activated SFK to inhibit the enzymatic activity of SFK (25). We examined whether LRT-SIFP was phosphorylated by SFK in MCF-7 cells. The six gradient fractions from MCF-7 cells expressing LRT-SIFP were immunoprecipitated with anti-phosphotyrosine antibody and were immunoblotted with anti-GFP antibody. LRT-SIFP was phosphorylated only in fraction 4, which is the DRM fraction, and this phosphorylation was blocked by treatment of cells with the specific SFK inhibitor PP2 for 2 h (Fig. 3D). In addition, we carried out immunoblotting of gradient fractions with anti–active Src antibody in the cells expressing LRT-SIFP. Compared with untransfected cells, the expression of LRT-SIFP significantly reduced the amount of active Src in fraction 4 (Fig. 3D). In contrast to active Src, the amount of Src in fraction 4 is not changed by the expression of LRT-SIFP (Fig. 3D). These results support that LRT-SIFP has a specific inhibitory effect on SFK activity in lipid rafts.

Physiologic effects of LRT-SIFP on tumor cell functions. Inhibition of SFK activity is known to suppress cell adhesion of many cell lines (22). We also confirmed that PP2, which ubiquitously inhibits SFK activity in cells, prevents cell adhesion of MCF-7 cells (Supplementary Fig. S1A). We then examined cell adhesion of MCF-7 cells expressing LRT-SIFP. The extent of adhesion of MCF-7 cells transfected with the SIFPs was measured and expressed in terms of the adhesion index, which is defined as the number of adhered fluorescence-positive cells divided by that of originally plated fluorescence-positive cells (see Materials and Methods). LRT-SIFP remarkably reduced the number of cells adhered to fibronectin, which is an integrin ligand (Fig. 4A). LRT-SIFP Del (=LRT-YFP), in which the peptide inhibitor of SFK is deleted (Fig. 3A), did not impair cell adhesion to fibronectin (Fig. 4A). LRT-SIFP Y6A contains the alanine-mutated peptide inhibitor MIYKYAF, whose inhibitory effect is reduced to about one-sixtieth (1/60; Fig. 3A; ref. 25). This LRT-SIFP Y6A did not suppress cell adhesion of MCF-7 cells (Fig. 4A). The reduction of cell adhesion by LRT-SIFP was not observed in the cells coexpressing wild-type (wt) Src and LRT-SIFP (Fig. 4A). In contrast to LRT-SIFP, SIFP, which is the YFP-fused peptide inhibitor of SFK lacking any targeting sequence (Fig. 3A), did not induce the reduction of MCF-7 cell adhesion (Fig. 4A). In addition, mLRT-SIFP, which has the mutated lipid raft–targeting sequence, did not inhibit MCF-7 cell adhesion (Figs. 3A and 4A). A control experiment was done to show that the effects of LRT-YFP and LRT-SIFP Y6A on cell adhesion can be modified by FBS stimulation. Cell adhesion of MCF-7 cells expressing LRT-SIFP and LRT-SIFP Y6A was inhibited in the absence of FBS stimulation (Supplementary Fig. S1B).

Figure 4.

LRT-SIFP inhibits cell adhesion of breast cancer MCF-7 and MDA-MB231 cells but does not inhibit cell adhesion of normal HEK293 cells. A, adhesion abilities of MCF-7 cells expressing GFP, LRT-YFP, LRT-SIFP, LRT-SIFP Y6A, and LRT-SIFP together with wt Src, SIFP, and mLRT-SIFP. LRT-SIFP reduced the adhesion ability of MCF-7 cells. The adhesion index is the number of the adhered fluorescence-positive cells divided by that of the originally plated fluorescence-positive cells (see Materials and Methods). Columns, mean of more than six independent experiments; bars, SE. ***, P < 0.001, significant difference in adhesion ability between cells expressing LRT-SIFP and controls (Student's t test). B, adhesion abilities of MDA-MB231 cells expressing LRT-YFP (open column) and LRT-SIFP (closed column). Columns, mean of six independent experiments; bars, SE. ***, P < 0.001, significant difference in adhesion ability between cells expressing LRT-SIFP and LRT-YFP (Student's t test). C, adhesion abilities of MDA-MB231 cells expressing LRT-YFP (open column) and LRT-SIFP (closed column). Columns, mean of six independent experiments; bars, SE.

Figure 4.

LRT-SIFP inhibits cell adhesion of breast cancer MCF-7 and MDA-MB231 cells but does not inhibit cell adhesion of normal HEK293 cells. A, adhesion abilities of MCF-7 cells expressing GFP, LRT-YFP, LRT-SIFP, LRT-SIFP Y6A, and LRT-SIFP together with wt Src, SIFP, and mLRT-SIFP. LRT-SIFP reduced the adhesion ability of MCF-7 cells. The adhesion index is the number of the adhered fluorescence-positive cells divided by that of the originally plated fluorescence-positive cells (see Materials and Methods). Columns, mean of more than six independent experiments; bars, SE. ***, P < 0.001, significant difference in adhesion ability between cells expressing LRT-SIFP and controls (Student's t test). B, adhesion abilities of MDA-MB231 cells expressing LRT-YFP (open column) and LRT-SIFP (closed column). Columns, mean of six independent experiments; bars, SE. ***, P < 0.001, significant difference in adhesion ability between cells expressing LRT-SIFP and LRT-YFP (Student's t test). C, adhesion abilities of MDA-MB231 cells expressing LRT-YFP (open column) and LRT-SIFP (closed column). Columns, mean of six independent experiments; bars, SE.

Close modal

We also examined whether LRT-SIFP suppresses cell adhesion of other cell lines. LRT-SIFP significantly inhibited cell adhesion in other breast cancer cell lines such as MDA-MB231 and MCF-7 (Fig. 4B). However, this inhibition of cell adhesion by LRT-SIFP was not observed in the human normal cell line HEK293 (Fig. 4C). These results indicate that the inhibitory effect of LRT-SIFP on cell adhesion is specific for tumor cell lines.

Another effect of inhibition of SFK activity is suppression of the tyrosine phosphorylation of cyclin D2, which is important for cell cycle progression at G1 phase (29). From cell cycle profiles obtained by flow cytometry with propidium iodide staining, we also confirmed that the 24-h incubation of MCF-7 cells with 5 μmol/L PP2 inhibits cell cycle progression (Supplementary Fig. S2). We next examined whether LRT-SIFP affects the cell cycle of MCF-7 cells. The cell cycle profiles of the YFP-positive cells expressing LRT-SIFPs were obtained by flow cytometry with propidium iodide staining (Fig. 5A). The cell cycle profiles of LRT-YFP, YFP, and mLRT-SIFP were used as a control for cell cycle progression of MCF-7 cells expressing LRT-SIFP, SIFP, and mLRT-SIFP, respectively (Fig. 5A,, C, and D). LRT-SIFP reduced the proportion of cells in G2-M phase to those in G1 phase, but LRT-SIFP Y6A did not reduce this proportion (Fig. 5A). As shown in Fig. 5B, LRT-SIFP significantly decreased the percentage of MCF-7 cells in G2-M phase as compared with LRT-YFP control (P < 0.001). The reduction by LRT-SIFP of the proportion of cells in G2-M phase to those in G1 phase was not observed in cells coexpressing wt Src and LRT-SIFP (Fig. 5A). In contrast to LRT-SIFP, the cell cycle profile of YFP-positive cells expressing SIFP, which is the YFP-fused peptide inhibitor of SFK alone, is similar to that of cells expressing YFP control (Fig. 5C). In addition, no difference in cell cycle profile was observed between mLRT-YFP control and mLRT-SIFP (Fig. 5D).

Figure 5.

LRT-SIFP inhibits cell cycle progression of MCF-7 cells. A, cell cycle profiles of MCF-7 cells expressing LRT-SIFP, LRT-SIFP Y6A, and LRT-YFP. The cell cycle profile of MCF-7 cells coexpressing wt Src and LRT-SIFP is also shown. Flow cytometry with use of propidium iodide staining shows the number of YFP-positive cells at the G1, S, and G2-M phases of the cell cycle. Only LRT-SIFP induced cell cycle arrest of MCF-7 cells. The cells were fixed and permeabilized with 70% ethanol for propidium iodide staining (see Materials and Methods). A, representative results of three independent experiments. B, percentages of MCF-7 cells in G2-M phase expressing LRT-YFP control and LRT-SIFP. The cell cycle data in (A) were used. The percentage was determined from the number of the YFP-positive cells in G2-M phase divided by that of the total YFP-positive cells. Columns, mean of three independent experiments; bars, SE. ***, P < 0.001, significant difference between cells expressing LRT-SIFP and LRT-YFP (Student's t test). C, cell cycle profiles of MCF-7 cells expressing SIFP and YFP. No significant difference in cell cycle profile was observed between SIFP and YFP. To avoid a drain of SIFP or YFP lacking the membrane-anchoring (lipid raft–targeting) sequence from the permeabilized cells, cells expressing SIFP or YFP were fixed with 1% PFA in PBS for 15 min at 4°C, and then permeabilized with Triton X-100 for propidium iodide staining (see Materials and Methods). Representative results of three independent measurements. D, cell cycle profiles of MCF-7 cells expressing mLRT-SIFP and mLRT-YFP. No difference in cell cycle profile was observed between mLRT-SIFP and mLRT-YFP. Representative results of three independent measurements. A to D, MCF-7 cells transfected with each construct were incubated for 24 h with medium containing 10% FBS before sample collection.

Figure 5.

LRT-SIFP inhibits cell cycle progression of MCF-7 cells. A, cell cycle profiles of MCF-7 cells expressing LRT-SIFP, LRT-SIFP Y6A, and LRT-YFP. The cell cycle profile of MCF-7 cells coexpressing wt Src and LRT-SIFP is also shown. Flow cytometry with use of propidium iodide staining shows the number of YFP-positive cells at the G1, S, and G2-M phases of the cell cycle. Only LRT-SIFP induced cell cycle arrest of MCF-7 cells. The cells were fixed and permeabilized with 70% ethanol for propidium iodide staining (see Materials and Methods). A, representative results of three independent experiments. B, percentages of MCF-7 cells in G2-M phase expressing LRT-YFP control and LRT-SIFP. The cell cycle data in (A) were used. The percentage was determined from the number of the YFP-positive cells in G2-M phase divided by that of the total YFP-positive cells. Columns, mean of three independent experiments; bars, SE. ***, P < 0.001, significant difference between cells expressing LRT-SIFP and LRT-YFP (Student's t test). C, cell cycle profiles of MCF-7 cells expressing SIFP and YFP. No significant difference in cell cycle profile was observed between SIFP and YFP. To avoid a drain of SIFP or YFP lacking the membrane-anchoring (lipid raft–targeting) sequence from the permeabilized cells, cells expressing SIFP or YFP were fixed with 1% PFA in PBS for 15 min at 4°C, and then permeabilized with Triton X-100 for propidium iodide staining (see Materials and Methods). Representative results of three independent measurements. D, cell cycle profiles of MCF-7 cells expressing mLRT-SIFP and mLRT-YFP. No difference in cell cycle profile was observed between mLRT-SIFP and mLRT-YFP. Representative results of three independent measurements. A to D, MCF-7 cells transfected with each construct were incubated for 24 h with medium containing 10% FBS before sample collection.

Close modal

We also examined the effect of LRT-SIFP on cell cycle progression of other cell lines. LRT-SIFP arrested cell cycle at G1 phase in MDA-MB231 cells as well as in MCF-7 cells (Figs. 5A and 6A). As shown in Fig. 6B, LRT-SIFP significantly decreased the percentage of MDA-MB231 cells in G2-M phase as compared with LRT-YFP control (P < 0.001). However, LRT-SIFP did not induce cell cycle arrest in normal HEK293 cells (Fig. 6C). These results indicate that the cell cycle of breast cancer cell lines is arrested by lipid raft–specific knockdown of SFK activity in a tumor-specific manner. LRT-SIFP was able to inhibit the SFK activity in lipid rafts, thereby preventing cell cycle progression and cell adhesion of breast cancer cells.

Figure 6.

LRT-SIFP inhibits cell cycle progression of MDA-MB231 cells but does not inhibit cell cycle progression of HEK293 cells. A, cell cycle profiles of MDA-MB231 cells expressing LRT-SIFP and LRT-YFP. Representative results of three independent experiments. B, percentages of MDA-MB231 cells in G2-M phase expressing LRT-YFP control and LRT-SIFP. The cell cycle data in (A) were used. The percentage was determined from the number of the YFP-positive cells in G2-M phase divided by that of the total YFP-positive cells. Columns, mean of three independent experiments; bars, SE. ***, P < 0.001, significant difference between cells expressing LRT-SIFP and LRT-YFP (Student's t test). C, cell cycle profiles of HEK293 cells expressing LRT-SIFP and LRT-YFP. A to C, cells transfected with each construct were incubated for 24 h with medium containing 10% FBS before sample collection.

Figure 6.

LRT-SIFP inhibits cell cycle progression of MDA-MB231 cells but does not inhibit cell cycle progression of HEK293 cells. A, cell cycle profiles of MDA-MB231 cells expressing LRT-SIFP and LRT-YFP. Representative results of three independent experiments. B, percentages of MDA-MB231 cells in G2-M phase expressing LRT-YFP control and LRT-SIFP. The cell cycle data in (A) were used. The percentage was determined from the number of the YFP-positive cells in G2-M phase divided by that of the total YFP-positive cells. Columns, mean of three independent experiments; bars, SE. ***, P < 0.001, significant difference between cells expressing LRT-SIFP and LRT-YFP (Student's t test). C, cell cycle profiles of HEK293 cells expressing LRT-SIFP and LRT-YFP. A to C, cells transfected with each construct were incubated for 24 h with medium containing 10% FBS before sample collection.

Close modal

From the pharmacologic point of view, peptide inhibitors of SFK have been developed by several groups (25, 30). These inhibitors are cell permeable and showed their high inhibitory effect on Src activity in in vitro experiments. However, none of these peptide inhibitors showed significant effects on cell functions in in vivo experiments (25). In addition, we showed that one of previously developed peptide inhibitors of SFK, MIYKYYF (=SIFP), did not inhibit cell adhesion and cell cycle progression of MCF-7 cells derived from human breast cancer. In contrast to these conventional peptide inhibitors, the present LRT-SIFP inhibits cell adhesion and cell cycle progression of breast cancer cell lines MCF-7 and MDA-MB231. These results conclude that the lipid raft–targeting of SIFP accelerates its inhibitory effect on SFK activity in lipid rafts, thereby causing the effective inhibition of tumor cell functions. Furthermore, LRT-SIFP did not show these inhibitory effects in normal HEK293 cells, suggesting that the present lipid raft–specific knockdown of SFK activity would potentially be useful for selective cancer therapy to prevent tumorigenesis and metastasis of breast cancer cells.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

Grant support: Japan Science and Technology Agency and Japan Society for the Promotion of Science.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

We thank Y. Imai, G. Wakamatsu, and M. Fujishiro for their help with the experiments and Dr. K. Owada for providing the anti–active Src antibody (clone 28).

1
Simons K, Toomre D. Lipid rafts and signal transduction.
Nat Rev Mol Cell Biol
2000
;
1
:
31
–9.
2
Cary LA, Cooper JA. Molecular switches in lipid rafts.
Nature
2000
;
404
:
945
–7.
3
Sargiacomo M, Sudol M, Tang Z, Lisanti MP. Signal transducing molecules and glycosyl-phosphatidylinositol-linked proteins form a caveolin-rich insoluble complex in MDCK cells.
J Cell Biol
1993
;
122
:
789
–807.
4
Liang X, Nazarian A, Erdjument-Bromage H, Bornmann W, Tempst P, Resh MD. Heterogeneous fatty acylation of Src family kinases with polyunsaturated fatty acids regulates raft localization and signal transduction.
J Biol Chem
2001
;
276
:
30987
–94.
5
Hitosugi T, Sasaki K, Sato M, Umezawa Y. Epidermal growth factor directs sex-specific steroid signaling through Src activation.
J Biol Chem
2007
;
282
:
10697
–706.
6
Sato M, Ozawa T, Inukai K, Asano T, Umezawa Y. Fluorescent indicators for imaging protein phosphorylation in single living cells.
Nat Biotechnol
2002
;
20
:
287
–94.
7
Kawabuchi M, Satomi Y, Takao T, et al. Transmembrane phosphoprotein Cbp regulates the activities of Src-family tyrosine kinases.
Nature
2000
;
404
:
999
–1003.
8
Takeuchi S, Takayama Y, Ogawa A, Tamura K, Okada M. Transmembrane phosphoprotein Cbp positively regulates the activity of the carboxyl-terminal Src kinase, Csk.
J Biol Chem
2000
;
275
:
29183
–6.
9
Ullman KS, Powers MA, Forbes DJ. Nuclear export receptors: from importin to exportin.
Cell
1997
;
90
:
967
–70.
10
Gagnoux-Palacios L, Dans M, van't Hof W, et al. Compartmentalization of integrin α6β4 signaling in lipid rafts.
J Cell Biol
2001
;
162
:
1189
–96.
11
Naal RMZG, Holowka EP, Baird B, Holowka D. Antigen-stimulated trafficking from the recycling compartment to the plasma membrane in RBL mast cells.
Traffic
2003
;
4
:
190
–200.
12
Mauro L, Sisci D, Bartucci M, et al. SHC-α5β1 integrin interactions regulate breast cancer cell adhesion and motility.
Exp Cell Res
1999
;
252
:
439
–48.
13
Perez-Martinez L, Jaworski DM. Tissue inhibitor of metalloproteinase-2 promotes neuronal differentiation by acting as an anti-mitogenic signal.
J Neurosci
2005
;
25
:
4917
–29.
14
Losel R, Wehling M. Nongenomic actions of steroid hormones.
Nat Rev Mol Cell Biol
2003
;
4
:
46
–56.
15
Steyer JA, Almers W. A real-time view of life within 100 nm of the plasma membrane.
Nat Rev Mol Cell Biol
2001
;
2
:
268
–75.
16
Harder T, Scheiffele P, Verkade P, Simons K. Lipid domain structure of the plasma membrane revealed by patching of membrane components.
J Cell Biol
1998
;
141
:
929
–42.
17
Young RM, Holowka D, Baird B. A lipid raft environment enhances Lyn kinase activity by protecting the active site tyrosine from dephosphorylation.
J Biol Chem
2003
;
278
:
20746
–52.
18
Lichtenberg D, Goni F, Heerklotz H. Detergent-resistant membranes should not be identified with membrane rafts.
Trends Biochem Sci
2006
;
30
:
430
–6.
19
Zajchowski LD, Robbins SM. Lipid rafts and little caves. Compartmentalized signalling in membrane microdomains.
Eur J Biochem
2002
;
269
:
737
–52.
20
Chapple JP, Grayson C, Hardcastle AJ, et al. Organization on the plasma membrane of the retinitis pigmentosa protein RP2: investigation of association with detergent-resistant membranes and polarized sorting.
Biochem J
2003
;
372
:
427
–33.
21
Kawakatsu H, Sakai T, Takagaki Y, et al. A new monoclonal antibody which selectively recognizes the active form of Src tyrosine kinase.
J Biol Chem
1996
;
271
:
5680
–5.
22
Playford MP, Schaller MD. The interplay between Src and integrins in normal and tumor biology.
Oncogene
2004
;
23
:
7928
–46.
23
Bromann PA, Korkaya H, Courtneidge SA. The interplay between Src family kinases and receptor tyrosine kinases.
Oncogene
2004
;
23
:
7957
–68.
24
Summy JM, Gallick GE. Src family kinases in tumor progression and metastasis.
Cancer Metastasis Rev
2003
;
22
:
337
–58.
25
Kamath JR, Liu R, Enstrom AM, Lou Q, Lam KS. Development and characterization of potent and specific peptide inhibitors of p60c-src protein tyrosine kinase using pseudosubstrate-based inhibitor design approach.
J Pept Res
2003
;
62
:
260
–8.
26
Zacharias DA, Violin JD, Newton AC, Tsien RY. Partitioning of lipid-modified monomeric GFPs into membrane microdomains of live cells.
Science
2002
;
296
:
913
–6.
27
Prior IA, Harding A, Yan J, Sluimer J, Parton RG, Hancock JF. GTP-dependent segregation of H-ras from lipid rafts is required for biological activity.
Nat Cell Biol
2001
;
3
:
368
–75.
28
Kenworthy AK, Nichols BJ, Remmert CL, et al. Dynamics of putative raft-associated proteins at the cell surface.
J Cell Biol
2001
;
165
:
735
–46.
29
Vercoutter-Edouart A, Lemoine J, Smart CE, et al. The mitogenic signaling pathway for fibroblast growth factor-2 involves the tyrosine phosphorylation of cyclin D2 in MCF-7 human breast cancer cells.
FEBS Lett
2000
;
478
:
209
–15.
30
Bogoyevitch MA, Barr RK, Ketterman AJ. Peptide inhibitors of protein kinases-discovery, characterisation and use.
Biochim Biophys Acta
2005
;
1754
:
79
–99.

Supplementary data