Abstract
Head and neck squamous cell carcinomas (HNSCCs) are frequently characterized by chemotherapy and radiation resistance and by overexpression of Bcl-Xl, an antiapoptotic member of the Bcl-2 protein family. In this report, we examined whether cell-permeable peptides derived from the BH3 domains of proapoptotic Bax, Bad, or Bak could be used to target Bcl-Xl and/or Bcl-2 in HNSCC cells and induce apoptotic death in these cells. To render the peptides cell-permeable, Antennapedia (Ant) or polyarginine (R8) peptide transduction domain was fused to the amino termini. Fluorescence microscopy of peptide-treated HNSCC cells revealed that the BH3 peptides colocalized with mitochondria, the site of Bcl-Xl and Bcl-2 expression. By contrast, a mutant peptide (BaxE BH3) that cannot bind Bcl-Xl or Bcl-2 was diffusely localized throughout the cytoplasm. Treatment of three HNSCC cell lines (1483, UM-22A, and UM-22B) with the wild-type BH3 peptides resulted in loss of viability and induction of apoptosis, as assessed by 3-(4,5-dimethythiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assays and annexin V staining. In general, Ant-conjugated peptides weremore potent than R8-conjugated peptides and Bad BH3 peptide was typically more potent than Bax BH3 or Bak BH3. Treatment of purified HNSCC mitochondria with BH3 peptides resulted in robust release of cytochrome c. Thus, the relative apoptosis resistance of HNSCC cells is not due to a deficit in this step of the intrinsic, mitochondrialmediated apoptosis pathway. We conclude that cellpermeable BH3 peptides can be used to target Bcl-Xl and/or Bcl-2 in HNSCC and that targeting of these proteins may have therapeutic value in the treatment of this disease.
Keywords: HNSCC, Bcl-Xl, Bcl-2, BH3 peptides, apoptosis
Introduction
Approximately 40,000 cases of head and neck squamous cell carcinoma (HNSCC) are diagnosed each year in the United States and roughly 500,000 cases are reported worldwide [1]. Standard treatment options for HNSCC include radiation, surgery, and chemotherapy. However, HNSCCs typically exhibit radiation and chemotherapy resistance, and 5-year survival rates have lingered around 50% for several decades [2,3]. Thus, there is a tremendous need to develop new therapeutic strategies for this disease.
The frequent resistance of HNSCC to radiation and chemotherapy is due, in large part, to aberrant inhibition of apoptosis in the tumor cells. A number of signaling proteins and pathways that contribute to cellular survival by blocking apoptosis have been shown to be overexpressed and/or hyperactivated in HNSCC, and include the epidermal growth factor receptor (EGFR) [4,5], Akt kinase [6,7], mammalian target of rapamycin [8], nuclear factor-κB (NF-κB) [9], and signal transducer and activator of transcription-3 (STAT3) [10–12]. Targeting EGFR with cetuximab monoclonal antibody, antisense oligonucleotides, dominant-negative mutants, or the tyrosine kinase inhibitors gefitinib and erlotinib serves to attenuate signaling downstream from the receptor and inhibits the proliferation and survival of HNSCC in vitro and in vivo [5,13–15]. Similarly, targeting of STAT3 or NF-κB with antisense, dominant-negatives, small interfering RNA, or a STAT3 decoy oligonucleotide promotes HNSCC cell apoptosis and enhances the sensitivities of these cells to cisplatin, tumor necrosis factor-α, histone deacetylase inhibitors, and radiation [10,16–22]. In addition, treatment with rapamycin or small molecule inhibitors of Akt has been shown to induce apoptosis and tumor regression in xenograft models of HNSCC [7,8,23]. The success of these molecular targeting strategies in preclinical models of HNSCC has raised hope for their successful application in patients.
HNSCC cell lines and primary specimens are also characterized by frequent overexpression of antiapoptotic members of the Bcl-2 protein family, including Bcl-2 and Bcl-Xl. In particular, Bcl-Xl is overexpressed in a large majority of HNSCC and is associated with chemoresistance in this disease [24]. The bcl-Xl gene is a known downstream target of STAT3 and NF-κB [25,26] and the overexpression of Bcl-Xl in HNSCC may be due to the aberrant activation of these transcription factors. Antisense oligonucleotides directed against Bcl-Xl or Bcl-2 mRNA have been shown to activate apoptosis and sensitize HNSCC cell lines to chemotherapy drugs [27].
The Bcl-Xl and Bcl-2 proteins act to inhibit chemotherapy-and radiation-induced apoptosis by regulating the intrinsic, mitochondrial-mediated pathway of apoptosis. Specifically, Bcl-Xl and Bcl-2 prevent chemotherapy- and radiation-induced release of cytochrome c from mitochondria, thereby preventing activation of the caspase protease cascade and apoptotic destruction of the cell [28]. Bcl-Xl and Bcl-2 exert these effects by binding to the BH3 domains of proapoptotic Bcl-2 family members such as Bax, Bad, and Bak, neutralizing the death-inducing effects of these proteins [28,29]. Recently, the small organic molecule gossypol has been shown to bind to the BH3-binding pocket of Bcl-Xl [30,31]. Treatment of HNSCC cells with (-)-gossypol, the biologically active enantiomer, has been shown to induce apoptosis and promote chemosensitization [32–34]. However, it remains uncertain whether the proapoptotic action of (-)-gossypol in HNSCC is due entirely to the inhibition of Bcl-Xl and/or Bcl-2 [35]. Thus, evaluation of other highly specific agents is needed to clearly validate the utility of Bcl-Xl/Bcl-2 targeting in HNSCC.
In the present study, we evaluated the ability of small peptides derived from the BH3 domains of Bax, Bad, and Bak to promote apoptosis signaling and death in HNSCC cells. Previous studies have shown that short peptides derived from the BH3 domains of proapoptotic proteins can disrupt the heterodimerization of Bcl-Xl and Bcl-2 with proapoptotic binding partners in a highly specific fashion, and induce apoptosis in leukemia and prostate cancer models [36–42]. Our results show that following addition to cells, the cell-permeable BH3 peptides localize to the mitochondria, the subcellular site of Bcl-Xl and Bcl-2 expression, supporting the ability of the peptides to specifically target Bcl-Xl and/or Bcl-2 in intact HNSCC cells. Peptide treatment resulted in disruption of heterodimerization interactions between pro- and antiapoptotic Bcl-2 family members. The cell-permeable peptides also stimulated caspase activation and apoptotic death in HNSCC cells, with Bad BH3 peptide exhibiting the most potent activity. Taken together, our results indicate the potential therapeutic benefit of targeting antiapoptotic members of the Bcl-2 protein family in HNSCC.
Materials and Methods
Cell Lines and Reagents
The UM-22A, UM-22B, and 1483 cell lines were derived from HNSCC and all are of human origin [43–46]. 1483 cells were provided by Dr. Gary Clayman (MD, Anderson Cancer Center, Houston, TX), and UM-22A and UM-22B were provided by Dr. Thomas Carey (University of Michigan, Ann Arbor, MI). Cells were cultured at 37°C and 5% CO2 in Dulbecco's modified Eagle's medium (DMEM; Mediatech, Herndon, VA) containing 10% heat-inactivated fetal bovine serum (FBS), 100 µg/ml penicillin-streptomycin, and 0.5 µg/ml amphotericin B. Jurkat T leukemic cells were grown at 37°C and 5% CO2 in RPMI medium (BioWhitaker, Walkersville, MD) containing 10% FBS, 100 µg/ml penicillin-streptomycin, and 0.5 µg/ml amphotericin B. Peptides were synthesized using a fluoren-9-ylmethoxycarbonyl synthesis protocol on a peptide synthesizer (Pioneer; Applied Biosystems, Foster City, CA) by the University of Pittsburgh Molecular Medicine Institute Peptide Synthesis Facility. Peptides were dissolved in DMSO and stored at -80°C. Hoechst stain solution was purchased from Sigma (St. Louis, MO) and MitoTracker Red fromMolecular Probes (Invitrogen, Carlsbad, CA). Streptavidin-fluorescein isothiocyanate (FITC) was from Molecular Probes as well. 3-(4,5-Dimethythiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) reagent was obtained from Promega (Madison, WI). Bio-Rad Protein Assay dye concentrate was purchased from Bio-Rad (Hercules, CA). Anti-Bcl-2 antibody was from DAKO (Carpinteria, CA) and anti-Bcl-Xl antibody was purchased from Santa Cruz Biotechnology (Santa Cruz, CA). Anti-poly(ADP)-ribose polymerase (PARP) antibody was from Cell Signaling Technology (Danvers, MA). Anti-β-actin antibody was from Sigma. Annexin V-FITC Apoptosis Detection Kits and anti-cytochrome c antibody were from BD Pharmingen (San Diego, CA).
Fluorescence Microscopy
Fluorescence microscopy was used to examine the uptake and subcellular distribution of BH3 peptides in HNSCC cells. Cells were seeded onto glass coverslips (Fisher Scientific, Pittsburgh, PA) at 1 × 104 cells per well in 24-well plates, grown overnight in 10% FBS/DMEM medium, then treated for varying lengths of time with biotinylated peptides (25 µM) or the peptide diluent DMSO (0.25%). Cells were then rinsed with cold PBS and incubated for 20 minutes at 37°C with 500 nM Mito Tracker Red to stainmitochondria. The cover-slips were then washed three times with 500 µl of cold PBS, and fixed with 3.7% paraformaldehyde for 30 minutes at 37°C. After fixation, cells were washed three times with 500 µl cold PBS, then permeabilized at room temperature with 2% paraformaldehyde containing 0.1%Triton X-100. Following another wash, the cells were incubated with 2 µg/ml streptavidin-FITC for 30 minutes at room temperature. Cells were also stained with 10 ng/ml Hoechst stain solution for 30 minutes at room temperature to identify nuclei. The coverslips were then transferred to slides and cells were visualized by fluorescence microscopy (Nikon Eclipse E800; Quantitative Imaging Corp., Burnaby, British Columbia, Canada).
Cell Viability Assays
For viability assays, cells were seeded at a density of 10,000 cells per well in 96-well plates, incubated overnight in 10% FBS/DMEM, washed with serum-free DMEM medium, then treated for 24 hours with varying doses of peptides in 100 µl of DMEM containing 1% FBS. Following treatment, cells were incubated for 2 hours at 37°C with 20 µl of MTS reagent. Absorbance was measured at 490-nm wavelength using a microplate reader (Bio-Tek Instruments, Inc., Winooski, VT). Data were analyzed by GraphPad PRISM version 4 software (San Diego, CA) to determine IC50 values.
Immunoblotting
Cells were harvested by centrifugation for 5 minutes at 4°C then washed once with cold PBS. The cell pellets were lysed for 10 minutes on ice in lysis buffer (50mMTris, pH 8.0, 150 mM NaCl, 0.1% SDS, and 1% NP-40) containing 3 µg/ml leupeptin, 20 µg/ml aprotinin, and 1.5 mM PMSF. The lysates were then pelleted at 11,000 × g for 2 minutes at 4°C and supernatants were transferred to new tubes. Protein concentrations were determined using Bio-Rad Protein Assay dye concentrate. For detection of Bcl-2 and Bcl-Xl, proteins (40 µg per lane) were electrophoresed on 12.5% SDS-PAGE gels. For detection of PARP cleavage products, proteins (60 µg per lane) were electrophoresed on 7.5% SDS-PAGE gels. Following electrophoresis, proteins were transferred to nitrocellulose membranes for 3 hours at 45 V. Membranes were blocked in 5% nonfat milk in TBST buffer (10 mM Tris pH 8.0, 150 mM NaCl, and 0.1% Tween 20) at room temperature for 1 hour, briefly washed with TBST buffer, and probed with primary antibody overnight at 4°C. After one 15-minute wash and three 5-minute washes in TBST buffer, the membranes were incubated with secondary antibody for 1 hour at room temperature. The membranes were then washed once with TBST buffer for 15 minutes, followed by three washes for 5 minutes each. The blots were developed using enhanced chemiluminescence reagent, according to the instructions of the manufacturer (PerkinElmer, Boston, MA).
Flow Cytometry Analysis
Externalization of phosphatidylserine was assessed using annexin V-FITC Apoptosis Detection Kits followed by flow cytometry. Cells were seeded at 1.8 × 106 cells per 100-mm dish, grown overnight in 10% FBS/DMEM, washed with serum-free DMEM, then treated for 4 hours with 25 µM of the peptides in 1% FBS/DMEM. Following incubation, cells were washed with cold PBS and removed from the plates by trypsinization. The resuspended cells were washed with cold PBS and stained with FITC-conjugated annexin V antibody and propidium iodide (PI) according to the instructions provided by the manufacturer (Roche Applied Science, Indianapolis, IN). Cells (50,000 per sample) were then subjected to flow cytometric analysis using a flow cytometer (Epics Coulter XL; Beckman-Coulter, Inc., Miami, FL) equipped with a 488-nm argon laser and Expo 32 software (Beckman-Coulter).
Mitochondria Isolation and Cytochrome c Release Assays
Mitochondria were isolated from healthy HNSCC cell lines and Jurkat cells growing in 10% FBS/DMEM. When the cells achieved approximately 80% confluence, 1 × 107 cells were harvested by centrifugation for 8 minutes at 4°C. The cells were washed once with cold PBS, centrifuged again, then resuspended in 100 µl of resuspension buffer (20 mM Hepes, pH 7.4, 10 mM KCl, 1.5 mM MgCl2, 1 mM EDTA, 250 mM sucrose, 1 mM DTT, 1.5 mM PMSF, 3 µg/ml leupeptin, and 20 µg/ml aprotinin). The resuspended cells were then homogenized using a type B tight-clearance dounce homogenizer (Wheaton, Millvale, NJ). The homogenized lysates were centrifuged at 11,000 × g for 30 minutes at 4°C to pellet mitochondria. The pellets were washed in 100 µl of resuspension buffer then centrifuged again at 11,000 × g for 30 minutes at 4°C. The mitochondria-containing pellets were resuspended in 100 µl of resuspension buffer and protein concentrations were determined using Bio-Rad Protein Assay dye concentrate. To perform cytochrome c release assays, resuspended mitochondria (40 µg) were incubated in the absence or presence of 25 µM of the BH3 peptides or DMSO (2%) for 1 hour at 37°C. Following incubation, the treated mitochondria were centrifuged at 11,000 × g for 30 minutes at 4°C and the supernatants were transferred to new tubes. The supernatants, containing proteins released from the mitochondria, were then subjected to a second centrifugation at 11,000 × g for 30 minutes at 4°C to eliminate potential contamination with intact mitochondria. Aliquots of the supernatants (50 µl) were then electrophoresed on 12.5% SDS-PAGE gels, transferred to nitrocellulose membranes, and subjected to immunoblotting with anti-cytochrome c antibody.
Results
Cell-Permeable Bax BH3 and Bad BH3 Peptides Localize to Mitochondria in HNSCC Cells
To determine whether peptides corresponding to the BH3 domains of proapoptotic proteins could be used to target Bcl-Xl and/or Bcl-2 in HNSCC cells, fluorescence microscopy was used to examine the subcellular localization of the peptides in treated cells. Bcl-Xl and Bcl-2 are known to be expressed on the outer membrane of mitochondria. Thus, agents that selectively bind and target Bcl-Xl and Bcl-2 would be expected to localize to mitochondria. Figure 1A depicts peptide localization in the HNSCC cell line UM-22B. The peptides were made cell-permeable by conjugating the 16-amino acid Antennapedia (Ant) peptide transduction domain [47] to the amino terminus of the 20-amino acid Bax BH3 peptide and the 21-amino acid Bad BH3 peptide (Table 1) [38,40]. As a control, Ant was also conjugated to the amino terminus of a mutant version of Bax BH3 peptide (termed BaxE), wherein a critical leucine residue in the wild-type peptide was changed to a glutamate residue [38,48]. In full-length Bax protein, mutation of this leucine residue to glutamate has been shown to abolish the ability of Bax to bind to antiapoptotic proteins and promote apoptosis [48]. All three peptides (Ant-Bax, Ant-BaxE, and Ant-Bad) were biotinylated, allowing fluorescent detection by streptavidin-FITC. Mitochondria were stained using MitoTracker Red dye and Hoechst staining was used to show nuclei. The UM-22B cells were treated for 2 or 6 hours with peptides (25 µM) or with 0.25% DMSO, the peptide diluent. As shown (Figure 1A), after 2 hours of treatment, all three peptides were detected in the cytoplasm of the treated cells. In the case of Ant-Bad, and particularly Ant-Bax, the cytoplasmic staining exhibited a punctate pattern similar to that observed for MitoTracker Red staining of mitochondria. Conversely, the mutant control peptide Ant-BaxE demonstrated diffuse cytoplasmic staining. When fluorescence from the peptide (green) and mitochondria (red) staining were merged, substantial yellow fluorescence was observed in cells treated with Ant-Bad and Ant-Bax. This indicated that the Ant-Bad and Ant-Bax peptides were localizing to the mitochondria, the site of Bcl-Xl and Bcl-2 expression. By contrast, no yellow fluorescence was observed in cells treated with Ant-BaxE, which is defective in its ability to bind to Bcl -Xl or Bcl-2. By 6 hours, robust levels of Ant-Bax and Ant-Bad were observed in the cytoplasm and yellow fluorescence indicative of peptide localization at the mitochondria was seen in the merged images. Moreover, cells treated with Ant-Bax or Ant-Bad for 6 hours exhibited altered morphology, with accompanying apoptotic debris, particularly in cells treated with Ant-Bad. Again, no mitochondrial localization (yellow fluorescence) of the control peptide Ant-BaxE was seen after 6 hours, and Ant-BaxE-treated cells did not exhibit any changes in morphology. Taken together, the fact that BH3 peptides capable of binding to Bcl-Xl and Bcl-2 were localized to the mitochondria, whereas a BH3 peptide that fails to bind Bcl-Xl and Bcl-2 did not localize to the mitochondria, indicates that the Ant-Bax and Ant-Bad peptides were targeting Bcl-Xl and Bcl-2 in the HNSCC cells. The disruption of cellular morphology and the appearance of apoptotic debris in the Ant-Bax- and Ant-Bad-treated cells further indicates that targeting of Bcl-Xl and/or Bcl-2 in HNSCC cells results in induction of apoptotic death, a finding explored in greater detail in subsequent experiments.
Figure 1.
Ant-Bax and Ant-Bad colocalize with mitochondria and disrupt Bcl-2/Bax complexes in UM-22B cells. (A) UM-22B cells were seeded onto glass coverslips at 1 × 104 cells per well (24-well plates), grown overnight, and treated with 25 µM biotinylated Ant-Bax, biotinylated Ant-Bad, biotinylated mutant peptide (Ant-BaxE), or with 0.25% DMSO (peptide solvent). After 2 and 6 hours of treatment, cells were incubated with 500 nM MitoTracker Red to stain mitochondria (red). Following fixation and permeabilization, cells were incubated with FITC-conjugated streptavidin to visualize peptide uptake (green). Cells were further incubated with Hoechst stain solution to stain nuclei (blue) and were then examined by fluorescence microscopy. A merged image (yellow) was used to detect colocalization of peptides (green) and mitochondria (red). The results demonstrate colocalization of Ant-Bax and Ant-Bad peptides, but not mutant Ant-BaxE, with mitochondria. Disruption of cellular morphology was also observed in cells treated with Ant-Bax (6 hours) or Ant-Bad (2 and 6 hours). (B)UM-22B cells were seeded at 1 × 106 cells per 100-mm dish, then treated for 2 hours in the absence or presence of 50 µM Ant-Bad, Ant-Bax, or Ant-BaxE mutant peptide, or 0.25% DMSO alone. Whole cell lysates (2 × 106 cells per lane) were prepared, then subjected to immunoprecipitation with anti-Bax polyclonal antibody, and followed by immunoblotting with anti-Bcl-2 monoclonal antibody.
Table 1.
Cell-Permeable BH3 Peptides.
Cell-Permeable Bax BH3 and Bad BH3 Peptides Disrupt Bcl-2/Bax Heterodimerization in Intact HNSCC Cells
To determine whether the cell-permeable BH3 peptides could disrupt physical interactions between pro- and antiapoptotic Bcl-2 family members in intact HNSCC cells, we performed coimmunoprecipitation experiments (Figure 1B). UM-22B cells were treated in the absence or presence of peptides, followed by immunoprecipitation with anti-Bax and immunoblotting with anti-Bcl-2 to detect Bcl-2/Bax heterodimerization complexes. Treatment of the cells with Ant-Bad or Ant-Bax resulted in a substantial decrease in the levels of intracellular Bcl-2/Bax, relative to untreated cells or cells treated with DMSO alone. By contrast, treatment with the mutant control peptide, Ant-BaxE, caused only a modest decline in Bcl-2/Bax heterodimer levels. These findings demonstrate that the wild-type BH3 peptides are capable of displacing proapoptotic proteins bound to antiapoptotic Bcl-2 family members in intact HNSCC cells.
Cell-Permeable BH3 Peptides Promote HNSCC Cell Death
We next examined whether cell-permeable BH3 peptides could induce cell death in HNSCC cell lines (Figure 2). Peptides corresponding to the BH3 domains of Bax, Bad, and Bak, as well as mutant control Bax (BaxE), were made cell-permeable by conjugating the Ant (Figure 2, A, C, and E) or polyarginine (R8; Figure 2, B, D, and F) peptide transduction domains to the amino termini of the peptides (Table 1). The cell-permeable peptides were then used to treat three different HNSCC cell lines, i.e., 1483, UM-22A, and UM-22B [43–45]. In this panel, UM-22A cells represent cells derived from a primary HNSCC tumor, whereas UM-22B cells were derived from a cervical lymph node metastasis in the same patient. 1483 cells were also derived from a primary tumor. Following treatment for 24 hours with varying doses of the peptides, MTS assays were performed, and IC50 values were determined (Table 2). As shown in Figure 2, the mutant control peptides (Ant-BaxE or R8-BaxE) exhibited little, if any, killing activity against the three HNSCC cell lines. By contrast, all three wild-type peptides demonstrated some level of killing activity. In general, peptides conjugated to Ant showed more potent killing activity than the R8-conjugated peptides. In addition, UM-22B cells exhibited greater resistance to the BH3 peptides than 1483 or UM-22A cells, consistent with the fact that UM-22B cells represent a more advanced, metastatic disease stage. In the case of 1483 cells, all three Ant-BH3 peptides promoted cell death, with IC50 values of 18.2 µM, 24.8 µM, and 32.9 µM for Ant-Bax, Ant-Bad, and Ant-Bak, respectively. In UM-22A cells, only Ant-Bad (IC50 = 18.3 µM) and Ant-Bax (IC50 = 45.5 µM) were effective at killing, whereas in UM-22B, only Ant-Bad (IC50 = 40.2 µM) exhibited activity. Similar results were obtained when Trypan Blue exclusion assays were employed (not shown).
Figure 2.
Antennapedia (Ant)- and polyarginine (R8)-conjugated BH3 domains of Bax, Bad, and Bak promote loss of HNSCC cell viability. 1483 (A and B), UM-22A (C and D), and UM-22B (E and F) cells were seeded into 96-well plates, grown overnight, then treated for 24 hours with varying doses of Ant- or R8-conjugated peptides in 1% FBS/DMEM medium. Following treatment, cells were analyzed using MTS assays. Each data point represents the average from triplicate wells and error bars represent standard deviations. Data were plotted as the percent of metabolic activity relative to untreated cells. IC50 values were calculated as described in the Materials and Methods section, and they are shown for Ant-conjugated peptides in Table 2. The experiments were performed three times, with similar results each time.
Table 2.
IC50 Values of Ant-Conjugated Peptides.
![]() |
To determine whether the differing sensitivities of the three HNSCC cell lines to the BH3 peptides may be due to different levels of expression of Bcl-Xl or Bcl-2, we performed immunoblotting with anti-Bcl-Xl or anti-Bcl-2 (Figure 3). Bcl-Xl has been reported to be frequently overexpressed in primary HNSCC specimens [24]. Consistent with this, all three cell lines were found to express abundant levels of the Bcl-Xl protein. 1483 cells had the lowest Bcl-Xl levels, correlating with the greater sensitivity of this cell line to the BH3 peptides. Bcl-2 levels varied among the three cell lines, with UM-22B cells exhibiting the highest expression. The high levels of Bcl-Xl and Bcl-2 found in UM-22B cells correlated with the greater resistance of these cells to the BH3 peptides. In addition, it is interesting to note that, whereas the Bcl-Xl levels in UM-22A and UM-22B were comparable, Bcl-2 expression was dramatically higher in UM-22B cells. This suggests, in this case, that conversion to a metastatic phenotype was associated with the acquisition of Bcl-2 overexpression status.
Figure 3.
Bcl-2 and Bcl-Xl expression levels in HNSCC cell lines. Whole cell protein extracts (40 µg per lane) from 1483, UM-22A, and UM-22B cells were subjected to immunoblotting with anti-Bcl-2 or anti-Bcl-Xl antibodies. Membranes were stripped and reprobed with anti-β-actin to demonstrate equal loading.
To determine whether the reduced killing activity of R8-conjugated peptides relative to Ant-conjugated peptides was due to reduced cellular uptake of the R8-conjugated peptides, we employed flow cytometry to assess peptide uptake (Figure W1). Cells were left untreated or were treated for 45 minutes with DMSO, biotinylated R8-Bax, or biotinylated Ant-Bax. Streptavidin-FITC and flow cytometry were then used to determine the percentage of cells that had taken up peptide. Surprisingly, whereas 50.4% of cells scored positive for uptake of R8-Bax in this experiment, only 23.0% of cells scored positive for Ant-Bax uptake (see Figure W1). The more efficient uptake, but reduced killing activity, of the R8-conjugated BH3 peptides suggests that these peptides may have reduced affinities for Bcl-Xl and/or Bcl-2 compared with the Ant-conjugated versions.
BH3 Peptides Induce HNSCC Apoptosis
Agents that directly target and inhibit antiapoptotic proteins such as Bcl-Xl and Bcl-2 might be expected to predispose a cell to apoptosis, or to directly activate apoptotic death. To determine whether the cell-permeable BH3 peptides activated an apoptotic form of cell death, HNSCC cell lines were exposed to peptides, then analyzed by flow cytometry for annexin V and PI staining. Annexin V staining serves as a measure of phosphatidylserine externalization, and cells that are annexin V+/PI- represent early apoptotic cells [49,50]. A representative experiment is shown in Figure 4A, in which UM-22A cells were treated with 25 µM peptide for 4 hours. As shown, the appearance of early apoptotic cells (annexin V+/PI-) is seen in cells treated with Ant-Bax, Ant-Bad, or Ant-Bak. By contrast, cells that were either untreated, or were treated with 0.25% DMSO or the control peptide Ant-BaxE (25 µM) showed no appreciable signs of apoptosis induction. After 4 hours of treatment, the Ant-Bad peptide produced the highest levels of apoptosis (31.7%), consistent with the results from MTS assays (Figure 2C). Similar results were seen in cells treated with peptides for 3 hours (data not shown). Longer treatment times were needed to observe high levels of apoptosis in Ant-Bax-treated cells (data not shown). However, Ant-BaxE mutant peptide failed to induce apoptosis even after prolonged incubation.
Figure 4.
Cell-permeable BH3 peptides activate apoptotic signaling and cell death. (A) UM-22A cells were seeded at 1.8 × 106 cells per 100-mm dish, grown overnight, treated for 4 hours with 0.25% DMSO or with 25 µM Ant-Bax, Ant-Bad, Ant-Bak, or Ant-BaxE peptides. Following incubation, cells were stained with annexin V-FITC and PI and subjected to flow cytometric analysis. (B) Cell-permeable BH3 peptides induce caspase protease activation in HNSCC cells. UM-22A cells were seeded at 1.8 × 106 cells per 100-mm dish, grown overnight, and then treated for 90 minutes with 0.5% DMSO or with 50 µM Ant-BaxE, Ant-Bax, Ant-Bad, or Ant-Bak. Following incubation, whole cell lysates were prepared and subjected to immunoblotting with anti-PARP antibody. A representative example of three independent experiments is shown.
The induction of apoptotic death is also characterized by the activation of caspase proteases, and the cleavage of caspase substrate proteins. Caspase-3, a major executioner caspase, is known to cleave PARP, a 116-kDa protein, to fragments of 89 and 24 kDa [51]. To determine whether treatment with the cell-permeable BH3 peptides resulted in caspase protease activation, treated HNSCC cells were analyzed by immunoblotting with anti-PARP antibody. A representative experiment using UM-22A cells treated for 90 minutes is shown in Figure 4B. Appearance of the 89-kDa PARP fragment was clearly visible in cells treated with Ant-Bad or Ant-Bak, although longer treatment times were needed to detect substantial PARP cleavage in Ant-Bax-treated cells. Taken together, our flow cytometric analyses and PARP cleavage analyses demonstrate that the cell-permeable BH3 peptides induce apoptotic cell death in HNSCC cells.
HNSCC Cells Are Not Defective in Mitochondrial-Mediated Apoptosis
We and others have previously reported that BH3 peptides potently kill chemotherapy-sensitive hematopoietic cell lines with IC50 values in the low micromolar range [39,40,52]. As shown in Figure 5A, when Jurkat T leukemic cells were treated for 48 hours with Ant-Bad or Ant-Bax, IC50 values of 3.8 µM and 5.6 µM, respectively, were observed. Much higher concentrations were needed to kill HNSCC cell lines (Figure 2 and Table 2). Because HNSCC cells are typically more resistant to chemotherapy drugs and because chemotherapy-induced apoptosis is known to be mediated by the mitochondrial or intrinsic apoptosis pathway [53], this raised the possibility that HNSCC cells may be defective in mitochondrial-mediated apoptosis signaling. To determine whether the reduced responsiveness of HNSCC cell lines to BH3 peptides was due to defective mitochondrial signaling, we isolated mitochondria from the three HNSCC cell lines and from Jurkat cells. The isolated mitochondria were then treated with peptides and the release of cytochrome c from mitochondria was assessed by immunoblotting of the supernatants (Figure 5B). The release of cytochrome c into cytosol represents a key step during the intrinsic, mitochondrial-mediated pathway of apoptosis [28,54]. As shown in Figure 5B, Ant-Bax, Ant-Bad, and Ant-Bak induced potent release of cytochrome c from the HNSCC mitochondria. By contrast, only negligible release was detected from HNSCC mitochondria treated with DMSO or the control peptide Ant-BaxE. These data were confirmed by immunoblotting of the mitochondrial pellets to show depletion of mitochondrial cytochrome c by the wild-type peptides (Figure 5B). Collectively, these results provide further support for the contention that the wild-type BH3 peptides directly target and inhibit Bcl-Xl and/or Bcl-2 on the surface of HNSCC mitochondria. In the case of isolated Jurkat mitochondria, the background levels of cytochrome c release detected in untreated, DMSO-treated, and Ant-BaxE-treated were significantly elevated. However, when shorter exposures of the gel were used to reduce the intensity of the background bands (compare 20-second exposure for Jurkat mitochondria with 1-minute exposure for HNSCC mitochondria), it can be seen that the induction of cytochrome c release by wild-type BH3 peptides is roughly comparable between Jurkat mitochondria and mitochondria from the HNSCC cell lines. This indicates that HNSCC cells are not defective in this key step in the intrinsic, mitochondrial-mediated apoptosis pathway.
Figure 5.
BH3 peptides induce comparable cytochrome c release from mitochondria isolated from chemosensitive Jurkat cells and chemoresistant HNSCC cells. (A) Cell-permeable BH3 peptides are potently active at killing Jurkat T leukemic cells. Jurkat cells were seeded at 1 × 105 cells/ml in 96-well plates in triplicate, grown overnight, and then treated with varying doses of Ant-Bad, Ant-Bax, or Ant-BaxE mutant peptide, as well as with Ant peptide alone. Following incubation, cells were analyzed using MTS assays as described in Figure 2. (B) Induction of cytochrome c release from mitochondria isolated from Jurkat, UM-22A, 1483, and UM-22B cells. Isolated mitochondria (40 µg per sample) were incubated for 1 hour at 37°C with 25 µM Ant-Bax, Ant-Bad, Ant-Bak, or the mutant control peptide, Ant-BaxE. Following incubation, the mitochondria were pelleted to obtain supernatants containing released cytochrome c. The supernatants and mitochondrial pellets were electrophoresed on 12.5% SDS-PAGE gels, transferred to nitrocellulose, and probed with anti-cytochrome c (Cyt. C). The experiment was performed three times, with similar results each time.
Discussion
HNSCC is a common human cancer with poor 5-year survival rates [1–3]. A major underlying problem in the treatment of HNSCC is the inherent resistance of the cancer cells to chemotherapy and radiation. This resistance is caused, in large part, by the aberrant inhibition of apoptosis signaling. In this report we sought to determine whether direct targeting of Bcl-Xl and/or Bcl-2 would promote apoptosis induction in HNSCC cells. Although previous reports have shown that (-)-gossypol, a small organic molecule that binds Bcl-Xl, promotes apoptosis in HNSCC cells [32–34], it has been difficult to validate Bcl-Xl and/or Bcl-2 as the primary targets of (-)-gossypol in intact HNSCC cells [35]. The BH3 peptides used in our studies offer the advantage that they can be labeled with biotin and be detected with streptavidin-FITC to demonstrate colocalization with the site of Bcl-Xl/Bcl-2 expression. In addition, BH3 peptides derived from proapoptotic proteins are highly specific for binding to antiapoptotic members of the Bcl-2 protein family [38,39,42,53,55–57]. Our results show that wild-type BH3 peptides colocalize with mitochondria in intact HNSCC cells, the site of Bcl-Xl and Bcl-2 expression (Figure 1A). The fact that a mutant BH3 peptide (Ant-BaxE) incapable of binding to Bcl-Xl or Bcl-2 no longer localized to mitochondria strongly supports the contention that the wild-type BH3 peptides directly targeted Bcl-Xl and/or Bcl-2 in intact HNSCC cells. This was supported by the finding that wild-type, and not mutant, peptides caused physical disruption of Bcl-2/Bax complexes in intact cells (Figure 1B). In addition, the wildtype Bax, Bad, and Bak BH3 peptides, and not the BaxE mutant peptide, were able to promote cytochrome c release from isolated HNSCC mitochondria (Figure 5). Together, these experiments indicate direct intracellular targeting of Bcl-Xl and/or Bcl-2 by the cell-permeable BH3 peptides.
Treatment of the three different HNSCC cell lines with the cell-permeable BH3 peptides resulted in activation of apoptosis signaling, as evidenced by phosphatidylserine externalization (annexin V staining) and PARP cleavage (Figure 4). Ultimately, loss of viability and cell death were observed (Figure 2). Thus, utilization of the BH3 peptides provided clear demonstration that direct targeting of Bcl-Xl and/or Bcl-2 in HNSCC cells leads to apoptosis induction. Comparison of the different peptides revealed that Ant-conjugated peptides were generally more efficacious at killing HNSCC cells than the R8-conjugated peptides. By contrast, R8-conjugated peptides appear to be taken up with slightly greater efficiency than the Ant-conjugated peptides. This suggests that conjugation to the Ant or R8 peptide transduction domains may differentially alter the affinities of the BH3 peptides for Bcl-Xl and/or Bcl-2.
We also observed that the cell-permeable Bad BH3 peptide was usually the most potent peptide for inducing apoptosis signaling and cell death. Prior studies have shown that full-length Bad protein and Bad BH3 peptide bind with higher affinity to Bcl-Xl than to Bcl-2 [58,59]. Because Bcl-Xl appears to be more highly overexpressed in the HNSCC cell lines we employed (Figure 3), an agent that exhibits preferential binding for Bcl-Xl might be predicted to exert more potent biological effects, as was observed in the case of Bad BH3. Other studies using in vitro binding assays have shown that Bad BH3 peptide is more efficacious than Bax BH3 peptide at disrupting interactions between Bcl-Xl and Bax proteins [38,40]. Because Bcl-Xl overexpression is more common than Bcl-2 overexpression in HNSCC primary specimens [24,60], it may be useful to focus on the Bad BH3 peptide for future efforts aimed at optimization and generation of derivatives or analogs.
The application of peptidic agents to the treatment of cancer is complicated by issues of size, stability, and uptake of the peptides or peptidomimetic derivatives. Nonetheless, our studies using BH3 peptides demonstrate that specific targeting of antiapoptotic Bcl-2 family members can be used to enhance the apoptotic sensitivities of HNSCC cells. Our findings support investigation in HNSCC of novel small molecule inhibitors of Bcl-Xl and/or Bcl-2, including agents such as ABT-737 and A-385358 which exhibit highly potent activities against solid tumor cell lines [61,62]. The eventual clinical application of highly specific and potent Bcl-Xl/Bcl-2 targeting agents may have significant therapeutic value in the treatment of chemotherapy- and radiation-resistant HNSCC.
Supplementary Material
Abbreviations
- Ant
Antennapedia
- DMEM
Dulbecco's modified Eagle's medium
- EGFR
epidermal growth factor receptor
- FITC
fluorescein isothiocyanate
- HNSCC
head and neck squamous cell carcinoma
- NF-κB
nuclear factor-kappaB
- PARP
poly(ADP)-ribose polymerase
- PI
propidium iodide
- R8
polyarginine
- STAT3
signal transducer and activator of transcription-3
Footnotes
This article refers to supplementary material, which is designated by Figure W1 and is available online at www.bcdecker.com.
This work was supported by a grant (R01-CA86980) from the National Institutes of Health and by a grant (P50-CA097190) from the University of Pittsburgh Specialized Program of Research Excellence (SPORE) in Head and Neck Cancer.
References
- 1.Jemal A, Siegel R, Ward E, Murray T, Xu J, Smigal C, Thun MJ. Cancer statistics, 2006. CA Cancer J Clin. 2006;56:106–130. doi: 10.3322/canjclin.56.2.106. [DOI] [PubMed] [Google Scholar]
- 2.Forastiere A, Koch W, Trotti A, Sidransky D. Head and neck cancer. New Eng J Med. 2001;345:1890–1900. doi: 10.1056/NEJMra001375. [DOI] [PubMed] [Google Scholar]
- 3.Mao L, Hong WK, Papadimitrakopoulou VA. Focus on head and neck cancer. Cancer Cell. 2004;5:311–316. doi: 10.1016/s1535-6108(04)00090-x. [DOI] [PubMed] [Google Scholar]
- 4.Rubin Grandis J, Melhem MF, Gooding WE, Day R, Holst VA, Wagener MM, Drenning SD, Tweardy DJ. Levels of TGF-alpha and EGFR protein in head and neck squamous cell carcinoma and patient survival. J Natl Cancer Inst. 1998;90:824–832. doi: 10.1093/jnci/90.11.824. [DOI] [PubMed] [Google Scholar]
- 5.Kalyankrishna S, Grandis JR. Epidermal growth factor receptor biology in head and neck cancer. J Clin Oncol. 2006;24:2666–2672. doi: 10.1200/JCO.2005.04.8306. [DOI] [PubMed] [Google Scholar]
- 6.Nakayama H, Ikebe T, Beppu M, Shirasuna K. High expression levels of nuclear factor kappaB, IkappaB kinase alpha and Akt kinase in squamous cell carcinoma of the oral cavity. Cancer. 2001;92:3037–3044. doi: 10.1002/1097-0142(20011215)92:12<3037::aid-cncr10171>3.0.co;2-#. [DOI] [PubMed] [Google Scholar]
- 7.Amornphimoltham P, Sriuranpong V, Patel V, Benavides F, Conti CJ, Sauk J, Sausville EA, Molinolo AA, Gutkind JS. Persistent activation of the Akt pathway in head and neck squamous cell carcinoma: a potential target for UCN-01. Clin Cancer Res. 2004;10:4029–4037. doi: 10.1158/1078-0432.CCR-03-0249. [DOI] [PubMed] [Google Scholar]
- 8.Amornphimoltham P, Patel V, Sodhi A, Nikitakis NG, Sauk JJ, Sausville EA, Molinolo AA, Gutkind JS. Mammalian target of rapamycin, a molecular target in squamous cell carcinomas of the head and neck. Cancer Res. 2005;65:9953–9961. doi: 10.1158/0008-5472.CAN-05-0921. [DOI] [PubMed] [Google Scholar]
- 9.Ondrey FG, Dong G, Sunwoo J, Chen Z, Wolf JS, Crowl-Bancroft CV, Mukaida N, Van Waes C. Constitutive activation of transcription factors NF-(kappa)B, AP-1, and NF- IL6 in human head and neck squamous cell carcinoma cell lines that express pro-inflammatory and pro-angiogenic cytokines. Mol Carcinog. 1999;26:119–129. doi: 10.1002/(sici)1098-2744(199910)26:2<119::aid-mc6>3.0.co;2-n. [DOI] [PubMed] [Google Scholar]
- 10.Grandis JR, Drenning SD, Zeng Q, Watkins SC, Melhem MF, Endo S, Johnson DE, Huang L, He Y, Kim JD. Constitutive activation of Stat3 signaling abrogates apoptosis in squamous cell carcinogenesis in vivo. Proc Natl Acad Sci USA. 2000;97:4227–4232. doi: 10.1073/pnas.97.8.4227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Masuda M, Suzui M, Yasumatu R, Nakashima T, Kuratomi Y, Azuma K, Tomita K, Komiyama S, Weinstein IB. Constitutive activation of signal transducers and activators of transcription 3 correlates with cyclin D1 overexpression and may provide a novel prognostic marker in head and neck squamous cell carcinoma. Cancer Res. 2002;62:3351–3355. [PubMed] [Google Scholar]
- 12.Sriuranpong V, Park JI, Amornphimoltham P, Patel V, Nelkin BD, Gutkind JS. Epidermal growth factor receptor-independent constitutive activation of STAT3 in head and neck squamous cell carcinoma is mediated by the autocrine/paracrine stimulation of the interleukin 6/gp130 cytokine system. Cancer Res. 2003;63:2948–2956. [PubMed] [Google Scholar]
- 13.Burtness B. The role of cetuximab in the treatment of squamous cell cancer of the head and neck. Expert Opin Biol Ther. 2005;5:1085–1093. doi: 10.1517/14712598.5.8.1085. [DOI] [PubMed] [Google Scholar]
- 14.Cohen EE. Role of epidermal growth factor receptor pathway - targeted therapy in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck. J Clin Oncol. 2006;24:2659–2665. doi: 10.1200/JCO.2005.05.4577. [DOI] [PubMed] [Google Scholar]
- 15.Cohen EE, Kane MA, List MA, Brockstein BE, Mehrotra B, Huo D, Mauer AM, Pierce C, Dekker A, Vokes EE. Phase II trial of gefitinib 250 mg daily in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck. Clin Cancer Res. 2005;11:8418–8424. doi: 10.1158/1078-0432.CCR-05-1247. [DOI] [PubMed] [Google Scholar]
- 16.Rubin Grandis J, Zeng Q, Drenning SD. Epidermal growth factor receptor -mediated STAT3 signaling blocks apoptosis in head and neck cancer. Laryngoscope. 2000;110:868–874. doi: 10.1097/00005537-200005000-00016. [DOI] [PubMed] [Google Scholar]
- 17.Leong PL, Andrews GA, Johnson DE, Dyer KF, Xi S, Mai JC, Robbins PD, Gadiparthi S, Burke NA, Watkins SF, et al. Targeted inhibition of STAT3 with a decoy oligonucleotide abrogates head and neck cancer cell growth. Proc Natl Acad Sci USA. 2003;100:4138–4143. doi: 10.1073/pnas.0534764100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Xi S, Gooding WE, Grandis JR. In vivo antitumor efficacy of STAT3 blockade using a transcription factor decoy approach: implications for cancer therapy. Oncogene. 2005;24:970–979. doi: 10.1038/sj.onc.1208316. [DOI] [PubMed] [Google Scholar]
- 19.Duffey DC, Chen Z, Dong G, Ondrey FG, Wolf JS, Brown K, Siebenlist U, Van Waes C. Expression of a dominant-negative mutant inhibitor-kappaBalpha of nuclear factor-kappaB in human head and neck squamous cell carcinoma inhibits survival, proinflammatory cytokine expression, and tumor growth in vivo. Cancer Res. 1999;59:3468–3474. [PubMed] [Google Scholar]
- 20.Duffey DC, Crowl-Bancroft CV, Chen Z, Ondrey FG, Nejad-Sattari M, Dong G, Van Waes C. Inhibition of transcription factor nuclear factor-kappaB by a mutant inhibitor-kappaBalpha attenuates resistance of human head and neck squamous cell carcinoma to TNF-alpha caspase-mediated cell death. Br J Cancer. 2000;83:1367–1374. doi: 10.1054/bjoc.2000.1423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Loercher A, Lee TL, Ricker JL, Howard A, Geoghegen J, Chen Z, Sunwoo JB, Sitcheran R, Chuang EY, Mitchell JB, et al. Nuclear factor-kappaB is an important modulator of the altered gene expression profile and malignant phenotype in squamous cell carcinoma. Cancer Res. 2004;64:6511–6523. doi: 10.1158/0008-5472.CAN-04-0852. [DOI] [PubMed] [Google Scholar]
- 22.Duan J, Friedman J, Nottingham L, Chen Z, Ara G, Van Waes C. Nuclear factor-kappaB p65 small interfering RNA or proteasome inhibitor bortezomib sensitizes head and neck squamous cell carcinomas to classic histone deacetylase inhibitors and novel histone deacetylase inhibitor PXD101. Mo Cancer Ther. 2007;6:37–50. doi: 10.1158/1535-7163.MCT-05-0285. [DOI] [PubMed] [Google Scholar]
- 23.Mandal M, Younes M, Swan EA, Jasser SA, Doan D, Yigitbasi O, McMurphey A, Ludwick J, El-Naggar AK, Bucana C, et al. The Akt inhibitor KP372-1 inhibits proliferation and induces apoptosis and anoikis in squamous cell carcinoma of the head and neck. Oral Oncol. 2006;42:430–439. doi: 10.1016/j.oraloncology.2005.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Trask DK, Wolf GT, Bradford CR, Fisher SG, Devaney K, Johnson M, Singleton T, Wicha M. Expression of Bcl-2 family proteins in advanced laryngeal squamous cell carcinoma: correlation with response to chemotherapy and organ preservation. Laryngoscope. 2002;112:638–644. doi: 10.1097/00005537-200204000-00009. [DOI] [PubMed] [Google Scholar]
- 25.Catlett-Falcone R, Landowski TH, Oshiro MM, Turkson J, Levitzki A, Savino R, Ciliberto G, Moscinski L, Fernandez-Luna JL, Nunez G, et al. Constitutive activation of Stat3 signaling confers resistance to apoptosis in human U266 myeloma cells. Immunity. 1999;10:105–115. doi: 10.1016/s1074-7613(00)80011-4. [DOI] [PubMed] [Google Scholar]
- 26.Chen C, Edelstein LC, Gelinas C. The Rel/NF-kappaB family directly activates expression of the apoptosis inhibitor Bcl-x(L) Mol Cell Biol. 2000;20:2687–2695. doi: 10.1128/mcb.20.8.2687-2695.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Sharma H, Sen S, Lo Muzio L, Mariggio A, Singh N. Antisense-mediated downregulation of anti-apoptotic proteins induces apoptosis and sensitizes head and neck squamous cell carcinoma cells to chemotherapy. Cancer Biol Ther. 2005;4:720–727. doi: 10.4161/cbt.4.7.1783. [DOI] [PubMed] [Google Scholar]
- 28.Danial NN, Korsmeyer SJ. Cell death: critical control points. Cell. 2004;116:205–219. doi: 10.1016/s0092-8674(04)00046-7. [DOI] [PubMed] [Google Scholar]
- 29.Cheng EH, Wei MC, Weiler S, Flavell RA, Mak TW, Lindsten T, Korsmeyer SJ. Bcl-2, Bcl-X(L) sequester BH3 domain-only molecules preventing BAX- and BAK-mediated mitochondrial apoptosis. Mol Cell. 2001;8:705–711. doi: 10.1016/s1097-2765(01)00320-3. [DOI] [PubMed] [Google Scholar]
- 30.Kitada S, Leone M, Sareth S, Zhai D, Reed JC, Pellecchia M. Discovery, characterization, and structure-activity relationships studies of proapoptotic polyphenols targeting B-cell lymphocyte/leukemia-2 proteins. J Med Chem. 2003;46:4259–4264. doi: 10.1021/jm030190z. [DOI] [PubMed] [Google Scholar]
- 31.Becattini B, Kitada S, Leone M, Monosov E, Chandler S, Zhai D, Kipps TJ, Reed JC, Pellecchia M. Rational design and real time, in-cell detection of the proapoptotic activity of a novel compound targeting Bcl-X(L) Chem Biol. 2004;11:389–395. doi: 10.1016/j.chembiol.2004.02.020. [DOI] [PubMed] [Google Scholar]
- 32.Oliver CL, Bauer JA, Wolter KG, Ubell ML, Narayan A, O'Connell KM, Fisher SG, Wang S, Wu X, Ji M, et al. In vitro effects of the BH3 mimetic, (-)-gossypol, on head and neck squamous cell carcinoma cells. Clin Cancer Res. 2004;10:7757–7763. doi: 10.1158/1078-0432.CCR-04-0551. [DOI] [PubMed] [Google Scholar]
- 33.Bauer JA, Trask DK, Kumar B, Los G, Castro J, Lee JS, Chen J, Wang S, Bradford CR, Carey TE. Reversal of cisplatin resistance with a BH3 mimetic, (-)-gossypol, in head and neck cancer cells: role of wild-type p53 and Bcl. Mol Cancer Ther. 2005;4:1096–1104. doi: 10.1158/1535-7163.MCT-05-0081. [DOI] [PubMed] [Google Scholar]
- 34.Wolter KG, Wang SJ, Henson BS, Wang S, Griffith KA, Kumar B, Chen J, Carey TE, Bradford CR, D'Silva NJ. (-)-Gossypol inhibits growth and promotes apoptosis of human head and neck squamous cell carcinoma in vivo. Neoplasia. 2006;8:163–172. doi: 10.1593/neo.05691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Baumgrass R, Weiwad M, Erdmann F, Liu JO, Wunderlich D, Grabley S, Fischer G. Reversible inhibition of calcineurin by the polyphenolic aldehyde gossypol. J Biol Chem. 2001;276:47914–47921. doi: 10.1074/jbc.M103273200. [DOI] [PubMed] [Google Scholar]
- 36.Diaz JL, Oltersdorf T, Horne W, McConnell M, Wilson G, Weeks S, Garcia T, Fritz LC. A common binding site mediates heterodimerization and homodimerization of Bcl-2 family members. J Biol Chem. 1997;272:11350–11355. doi: 10.1074/jbc.272.17.11350. [DOI] [PubMed] [Google Scholar]
- 37.Finnegan NM, Curtin JF, Prevost G, Morgan B, Cotter TG. Induction of apoptosis in prostate carcinoma cells by BH3 peptides which inhibit Bak/Bcl-2 interactions. Br J Cancer. 2001;85:115–121. doi: 10.1054/bjoc.2001.1850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Shangary S, Johnson DE. Peptides derived from BH3 domains of Bcl-2 family members: a comparative analysis of inhibition of Bcl-2, Bcl-x(L) and Bax oligomerization, induction of cytochrome c release, and activation of cell death. Biochemistry. 2002;41:9485–9495. doi: 10.1021/bi025605h. [DOI] [PubMed] [Google Scholar]
- 39.Letai A, Bassik MC, Walensky LD, Sorcinelli MD, Weiler S, Korsmeyer SJ. Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell. 2002;2:183–192. doi: 10.1016/s1535-6108(02)00127-7. [DOI] [PubMed] [Google Scholar]
- 40.Shangary S, Oliver CL, Tillman TS, Cascio M, Johnson DE. Sequence and helicity requirements for the proapoptotic activity of Bax BH3 peptides. Mol Cancer Ther. 2004;3:1343–1354. [PubMed] [Google Scholar]
- 41.Moreau C, Cartron PF, Hunt A, Meflah K, Green DR, Evan G, Vallette FM, Juin P. Minimal BH3 peptides promote cell death by antagonizing anti-apoptotic proteins. J Biol Chem. 2003;278:19426–19435. doi: 10.1074/jbc.M209472200. [DOI] [PubMed] [Google Scholar]
- 42.Willis SN, Fletcher JI, Kaufmann T, van Delft MF, Chen L, Czabotar PE, Ierino H, Lee EF, Fairlie WD, Bouillet P, et al. Apoptosis initiated when BH3 ligands engage multiple Bcl-2 homologs, not Bax or Bak. Science. 2007;315:856–859. doi: 10.1126/science.1133289. [DOI] [PubMed] [Google Scholar]
- 43.Krause CJ, Carey TE, Ott RW, Hurbis C, McClatchey KD, Regezi JA. Human squamous cell carcinoma. Establishment and characterization of new permanent cell lines. Arch Otolaryngol. 1981;107:703–710. doi: 10.1001/archotol.1981.00790470051012. [DOI] [PubMed] [Google Scholar]
- 44.Sacks PG, Parnes SM, Gallick GE, Mansouri Z, Lichtner R, Satya-Prakash KL, Pathak S, Parsons DF. Establishment and characterization of two new squamous cell carcinoma cell lines derived from tumors of the head and neck. Cancer Res. 1988;48:2858–2866. [PubMed] [Google Scholar]
- 45.Grenman R, Carey TE, McClatchey KD, Wagner JG, Pekkola-Heino K, Schwartz DR, Wolf GT, Lacivita LP, Ho L, Baker SR, et al. In vitro radiation resistance among cell lines established from patients with squamous cell carcinoma of the head and neck. Cancer. 1991;67:2741–2747. doi: 10.1002/1097-0142(19910601)67:11<2741::aid-cncr2820671105>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
- 46.Lin CJ, Grandis JR, Carey TE, Gollin SM, Whiteside TL, Koch WM, Ferris RL, Lai SY. Head and neck squamous cell carcinoma cell lines: established models and rationale for selection. Head Neck. 2007;29:163–188. doi: 10.1002/hed.20478. [DOI] [PubMed] [Google Scholar]
- 47.Derossi D, Joliot AH, Chassaing G, Prochiantz A. The third helix of the Antennapedia homeodomain translocates through biological membranes. J Biol Chem. 1994;269:10444–10450. [PubMed] [Google Scholar]
- 48.Wang K, Gross A, Waksman G, Korsmeyer SJ. Mutagenesis of the BH3 domain of BAX identifies residues critical for dimerization and killing. Mol Cell Biol. 1998;18:6083–6089. doi: 10.1128/mcb.18.10.6083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Fadok VA, Voelker DR, Campbell PA, Cohen JJ, Bratton DL, Henson PM. Exposure of phosphatidylserine on the surface of apoptotic lymphocytes triggers specific recognition and removal by macrophages. J Immunol. 1992;148:2207–2216. [PubMed] [Google Scholar]
- 50.Martin SJ, Reutelingsperger CP, McGahon AJ, Rader JA, van Schie RC, LaFace DM, Green DR. Early redistribution of plasma membrane phosphatidylserine is a general feature of apoptosis regardless of the initiating stimulus: inhibition by overexpression of Bcl-2 and Abl. J Exp Med. 1995;182:1545–1556. doi: 10.1084/jem.182.5.1545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kaufmann SH, Desnoyers S, Ottaviano Y, Davidson NE, Poirier GG. Specific proteolytic cleavage of poly(ADP-ribose) polymerase: an early marker of chemotherapy-induced apoptosis. Cancer Res. 1993;53:3976–3985. [PubMed] [Google Scholar]
- 52.Wang JL, Zhang ZJ, Choksi S, Shan S, Lu Z, Croce CM, Alnemri ES, Korngold R, Huang Z. Cell permeable Bcl-2 binding peptides: a chemical approach to apoptosis induction in tumor cells. Cancer Res. 2000;60:1498–1502. [PubMed] [Google Scholar]
- 53.Shangary S, Johnson DE. Recent advances in the development of anticancer agents targeting cell death inhibitors in the Bcl-2 protein family. Leukemia. 2003;17:1470–1481. doi: 10.1038/sj.leu.2403029. [DOI] [PubMed] [Google Scholar]
- 54.Liu X, Kim CN, Yang J, Jemmerson R, Wang X. Induction of apoptotic program in cell-free extracts: requirement for dATP and cytochrome c. Cell. 1996;86:147–157. doi: 10.1016/s0092-8674(00)80085-9. [DOI] [PubMed] [Google Scholar]
- 55.Chen L, Willis SN, Wei A, Smith BJ, Fletcher JI, Hinds MG, Colman PM, Day CL, Adams JM, Huang DC. Differential targeting of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apoptotic function. Mol Cell. 2005;17:393–403. doi: 10.1016/j.molcel.2004.12.030. [DOI] [PubMed] [Google Scholar]
- 56.Kuwana T, Bouchier-Hayes L, Chipuk JE, Bonzon C, Sullivan BA, Green DR, Newmeyer DD. BH3 domains of BH3-only proteins differentially regulate Bax-mediated mitochondrial membrane permeabilization both directly and indirectly. Mol Cell. 2005;17:525–535. doi: 10.1016/j.molcel.2005.02.003. [DOI] [PubMed] [Google Scholar]
- 57.Certo M, Del Gaizo Moore V, Nishino M, Wei G, Korsmeyer S, Armstrong SA, Letai A. Mitochondria primed by death signals determine cellular addiction to antiapoptotic Bcl-2 family members. Cancer Cell. 2006;9:351–365. doi: 10.1016/j.ccr.2006.03.027. [DOI] [PubMed] [Google Scholar]
- 58.Yang E, Zha J, Jockel J, Boise LH, Thompson CB, Korsmeyer SJ. Bad, a heterodimeric partner for Bcl-Xl and Bcl-2, displaces Bax and promotes cell death. Cell. 1995;80:285–291. doi: 10.1016/0092-8674(95)90411-5. [DOI] [PubMed] [Google Scholar]
- 59.Ottilie S, Diaz JL, Horne W, Chang J, Wang Y, Wilson G, Chang S, Weeks S, Fritz LC, Oltersdorf T. Dimerization properties of human BAD Identification of a BH-3 domain and analysis of its binding to mutant Bcl-2 and Bcl-Xl proteins. J Biol Chem. 1997;272:30866–30872. doi: 10.1074/jbc.272.49.30866. [DOI] [PubMed] [Google Scholar]
- 60.Pena JC, Thompson CB, Recant W, Vokes EE, Rudin CM. Bcl-Xl and Bcl-2 expression in squamous cell carcinoma of the head and neck. Cancer. 1999;85:164–170. doi: 10.1002/(sici)1097-0142(19990101)85:1<164::aid-cncr23>3.0.co;2-q. [DOI] [PubMed] [Google Scholar]
- 61.Oltersdorf T, Elmore SW, Shoemaker AR, Armstrong RC, Augeri DJ, Belli BA, Bruncko M, Deckwerth TL, Dinges J, Hajduk PJ, et al. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature. 2005;435:677–681. doi: 10.1038/nature03579. [DOI] [PubMed] [Google Scholar]
- 62.Shoemaker AR, Oleksijew A, Bauch J, Belli BA, Borre T, Bruncko M, Deckwirth T, Frost DJ, Jarvis K, Joseph MK, et al. A smallmolecule inhibitor of Bcl-Xl potentiates the activity of cytotoxic drugs in vitro and in vivo. Cancer Res. 2006;66:8731–8739. doi: 10.1158/0008-5472.CAN-06-0367. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.