Purpose: Antiangiogenic therapy is considered as an effective strategy for controlling the growth and metastasis of tumors. Among a myriad of biological activities described for xanthone derivatives, the anticancer activity is quite remarkable, but the molecular mechanism is not clearly resolved. In the present study, we investigated the antiangiogenic mechanism of 3,6-di(2,3-epoxypropoxy)xanthone (EPOX), a novel Mcl-1 targeting drug.

Experimental Design: To evaluate the antiangiogenic activity of EPOX, we did cell viability, cell cycle, tube formation assay in vitro, and Matrigel plug assay in vivo. To evaluate the effect of EPOX on the endothelial signaling pathway, we did immunoblotting, immunoprecipitation, and immunofluorescence analysis. Intracellular glutathione levels were determined with the use of monochlorobimane, a glutathione-specific probe.

Results: EPOX induced endothelial cell apoptosis in association with proteasome-dependent Mcl-1 degradation. Down-regulation of Mcl-1 resulted in an increase in Mcl-1–free Bim, activation of Bax, and then signaling of mitochondria-mediated apoptosis. Additionally, glutathione depletion and extracellular signal-regulated kinase (ERK) inactivation was observed in EPOX-treated cells. Glutathione supplementation reversed the inhibitory effects of EPOX on ERK, which increases the phosphorylation of Mcl-1 at T163. Overexpression of mitogen-activated protein/ERK kinase (MEK) partially reversed the effect of EPOX on Mcl-1 dephosphorylation, ubiquitination, and degradation, further implicating ERK in the regulation of Mcl-1 stability.

Conclusions: This study provides evidence that EPOX induces glutathione depletion, ERK inactivation, and Mcl-1 degradation on endothelial cells, which leads to inhibition of angiogenesis. Our results suggest that EPOX is a novel antiangiogenic agent, making it a promising lead compound for further development in the treatment of angiogenesis-related pathologies.

Translational Relevance

Although some antiangiogenic drugs have been approved in the treatment of cancer, the survival benefits of such drugs are relatively modest. Finding new drugs and understanding the mechanism governing angiogenesis are therefore essential for therapeutic improvements. We found that 3,6-di(2,3-epoxypropoxy)xanthone (EPOX) was the most effective in inhibiting endothelial cell proliferation among a series of xanthone derivatives. The antiangiogenic actions of EPOX are proposed to involve the inhibition of extracellular signal-regulated kinase in response to glutathione depletion leading to the degradation of Mcl-1, and then the stimulation of mitochondria-mediated apoptosis in response to an increase in Mcl-1–free Bim. Our study clearly shows the mechanism of EPOX and represents the most thorough investigation to date of the biological consequence of Mcl-1 inhibition in endothelial cells. Furthermore, we proposed that therapies designed to manipulate reduced glutathione content, the extracellular signal-regulated kinase/Mcl-1 pathway, and crosstalk among Bcl-2 members could be a new direction for antiangiogenic therapy.

Angiogenesis, namely neovascularization from preexisting vasculature, is necessary for continued growth and metastasis of tumors (1). Angiogenesis-related molecules, such as vascular endothelial growth factor and vascular endothelial growth factor receptor, have been suggested to be ideal targets, and a large number of inhibitors have been developed. Apart from angiogenesis inhibitors, so-called vascular disrupting agents targeted at the established tumor vasculature are currently being investigated in clinical trial. Recently, we revealed that an endostatin-cytosine deaminase fusion protein possesses the tumor vessel–targeting property of endostatin to allow selective tumor-killing effect of cytosine deaminase, producing potent antitumor activity (2).

Endothelial cell apoptosis is intimately involved in the sculpting of blood vessels (3). Natural inhibitors of angiogenesis, such as endostatin, angiostatin, and thrombospondin-1, act in part through selective induction of endothelial cell apoptosis to promote active vessel regression (4). Metronomic chemotherapy, defined as the frequent administration of chemotherapeutic drugs at doses significantly below the maximally tolerated doses, continuously exposes tumor endothelial cells to these drugs. During such therapy, endothelial cell apoptosis precedes tumor cell apoptosis (5). In preclinical studies, metronomic chemotherapy was found effective for managing tumors with cancer cells that had developed resistance to the same chemotherapeutic agents (6). According to current laboratory and clinical findings, inducing apoptosis in tumor endothelial cells contributes to the antitumor efficacy of antiangiogenic and cytotoxic drugs (7).

Apoptosis is characterized by a variety of biochemical and morphologic alterations, including caspase activation, mitochondrial depolarization, chromatin condensation, cell shrinkage, and formation of apoptotic bodies (8). Intracellular redox status is recognized to play an important role in the induction of cell apoptosis. Glutathione (GSH), which accounts for >90% of cellular nonprotein thiols, is critical to maintaining cellular redox balance (9). Therefore, compounds that block the effects of GSH possess the potential to induce cancer cell apoptosis or to reverse drug resistance (10).

Xanthone compounds, secondary metabolites derived from higher plants and microorganisms, have diverse biological properties, including tuberculostatic, antithrombotic, antiallergic, and anticancer activities (11). The emerged “hit” xanthone derivatives, such as 5,6-dimethylxanthenone-4-acetic acid (DMXAA), psorospermin, mangiferin, norathyriol, mangostins, and AH6809, constitute excellent starting materials to leads development in the field of cancer. DMXAA has completed phase II testing (12) and is currently undergoing phase III evaluation in non–small cell lung cancer (ATTRACT-1[Antivascular Targeted Therapy: Researching ASA404 in Cancer Treatment] trial). DMXAA induces apoptosis of tumor vascular endothelial cells and cytokine production, leading to tumor vascular collapse (13). As we previously reported, a series of oxygenated xanthones and [3-(dialkylamino)-2-hydroxypropoxy]xanthones have been prepared and tested for anticancer activity (14). 2,6-Di(2,3- epoxypropoxy)xanthone was chosen for further study, and the mechanism of action is proposed to involve a selective down- regulation of Ras (15). After examining the antiproliferation effects of xanthone derivatives on human umbilical vascular endothelial cells (HUVEC), structure-activity analysis indicated epoxidation of the hydroxyxanthone increased cytotoxicity, but ring opening of the epoxide group with dialkylamine did not enhance the antiangiogeneic activity (Supplementary Fig. S1A). 3,6-Di(2,3-epoxypropoxy)xanthone (EPOX; Supplementary Fig. S1B) was found to be the most effective. The present study was therefore conducted to elucidate the mechanism whereby EPOX exerts its apoptotic effects in endothelial cells.

Reagents

Xanthone derivatives were provided by Chun-Nan Lin (Kaohsiung Medical University, Kaohsiung, Taiwan). EPOX was synthesized by a method described previously (14). Briefly, 3,6-dihydroxyxanthone was reacted with one equivalent of NaOH in aqueous 2-propanol and an excess of epichlorohydrin to yield the EPOX. The melting point of EPOX is 187°C to 188°C. Anti-Bax 6A7 and all chemicals, unless specifically mentioned, were ordered from Sigma. 4,6-Diamidino-2-phenylindole, anti-HA, were purchased from Roche Diagnostics Corp. Rhodamine 123, anti–complex IV, and MitoTracker were obtained from Molecular Probes. The caspase inhibitor Z-VAD-fmk was purchased from Alexis. Caspase-3 antibody was from Imgenex. Antibodies against caspase-8, caspase-9, cleaved caspase-3, cleaved poly(ADP-ribose) polymerase, phospho and total extracellular signal-regulated kinase (ERK), phospho and total GSK-3β, and phospho and total c-Jun-NH2-kinase (JNK) were purchased from Cell Signaling Technologies. Cytochrome c and Mcl-1 antibodies came from BD Pharmingen. Antibodies against Bcl-2, Bcl-xL, Bax, Bak, Bim, actin, and α-tubulin were obtained from Santa Cruz Biotechnology. Antibodies against phosphorylated Thr 163 of Mcl-1 were generated by Bethyl Laboratories, Inc. (16). pHA-hMcl-1 was kindly provided by Hsin-Fang Yan-Yen (Academia Sinica, Taipei, Taiwan). Constitutively active mitogen-activated protein (MAP)/ERK kinase (MEK) was a gift from Pier Paolo Pandolfi (Harvard Medical School, Boston, MA).

Cell culture and transfection

HUVECs were obtained from human umbilical cord veins and cultured as previously described (17). The endothelial-like EAhy926 cells were maintained in DMEM supplemented with 100 μmol/L sodium hypoxanthine, 0.4 μmol/L aminopterin, and 16 μmol/L thymidine. These cells retain the properties of endothelial cells, including expression of factor VIII–related antigen and tube-forming activity (18). Cells were transfected with Lipofectamine 2000 (Invitrogen) according to the manufacturer's protocol. Small interfering RNA (SiRNA)-mediated down-regulation of Bim was done by transfecting SMARTpool specific or nonspecific control double-stranded RNA oligonucleotides (Dharmacon). For the EPOX treatment, HUVECs were seeded and then incubated with 3 μmol/L EPOX unless specifically mentioned.

Evaluation of cell viability and identification of apoptosis

The percentage of surviving cells after treatment was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay (MTT) (19). Cell proliferation was determined with the use of a bromodeoxyuridine labeling and detection kit (Amersham Biosciences) following manufacturer's guidelines. Cell cycle distribution was assessed by a flow cytometry assay as described (19). For the morphologic examination, cells were examined under a phase contrast microscope (Nikon). To identify apoptotic cells, the cells were inspected by 4′,6-diamidino-2-phenylindole staining (DAPI) as previously described (19). For a quantitative assessment of oligonucleosomal DNA fragmentation, apoptosis was detected through a Cell Death ELISAPLUS kit (Roche Molecular Biochemicals) according to the manufacturer's protocol. Measurement of mitochondrial membrane potential was determined as detailed elsewhere (20). The caspase colorimetric assay kits specific for caspase-3, caspase-8, or caspase-9 (Biovision) were used to detect caspase activation.

Tube formation assay

Tube formation assay was done as previously described (2). In brief, 3 × 104 endothelial cells were seeded on a layer of polymerized Matrigel (BD Biosciences) in a 96-well plate. All assays were done in triplicate. Cells were incubated for 6 h and viewed under a microscope. Tube formation was quantified by measuring the long axis of each tube in three random fields per well with the use of Image-Pro Plus software.

In vivo Matrigel plug assay

In vivo Matrigel plug assay was done as previously described (17). In brief, C57BL/6J mice were given s.c. injections of 0.5 mL of Matrigel (BD Biosciences) mixed with endothelial growth medium 2 (EGM2; Clonetics) growth medium. Matrigel pellets were harvested on day 7 and were processed for Masson's trichrome staining. Hemoglobin was measured as an indication of blood vessel formation with the use of the Drabkin method (Sigma-Aldrich).

Tumor xenografts

For HCT-116 cancer cell xenograft experiments, male athymic nude mice were inoculated s.c. with 107 cells per mouse. When the tumors reached the average volume of 50 mm3, the mice were randomized into two groups (n = 5 each). EPOX oral treatment at a dose of 50 mg/kg was given daily. Tumors were measured with a caliper every 3 to 4 d, and their volumes were calculated with the formula: 0.5 L × W2, where L and W are the long and short diameters of the tumor, respectively. For histologic analysis, the xenograft tissue samples were stained with CD31 antibody to permit determination of the number of blood vessels. All animal experiments were done in accordance with institutional guidelines for animal welfare.

Immunoblotting and immunoprecipitation

Immunoblotting and immunoprecipitation were done essentially as previously described (21). For quantification of the western analysis, the density of each band was quantified by ImageQuant software and indicated below the blots. Enriched mitochondrial fraction was isolated according to protocols from Yamaguchi et al. (22). For detection of Bax conformational change, cells were lysed with Chaps lysis buffer (10 mmol/L HEPES, pH 7.4, 150 mmol/L NaCl, 1% Chaps) and subjected to immunoprecipitation with anti-Bax 6A7 as described (22).

Detection of glutathione by monochlorobimane

The content of glutathione was determined with the use of monochlorobimane, a permeable probe that forms blue fluorescent adducts with the reduced form of GSH. To determine the GSH level, after treatment cells in 96-well plates were incubated with 100 μmol/L monochlorobimane for 30 min in the dark. Then, plates were read on a microplate reading fluorometer with excitation and emission wavelengths of 390 and 460 nm, respectively. For imaging the intracellular GSH, cells seeded into a 4-well slide chamber were incubated with monochlorobimane as described above. Cells were fixed for detecting the fluorescence images with a fluorescence microscope (Nikon).

Immunofluorescence analysis

Cells were treated with EPOX for 6 h. After incubation with 25 nmol/L MitoTracker Red CMXRos (Molecular Probes) for 30 min, cells were fixed with ice-cold methanol for 15 min at 4°C and blocked for 30 min with 3% bovine serum albumin. Cells were then incubated with anti-Bax (1:100) overnight at 4°C, followed by incubation at 37°C for 1 h with FITC-conjugated secondary antibody. The slide chamber was mounted onto glass slides for microscopic observation with a Leica TCS SP2 confocal spectral microscope.

Statistical analysis

Each experiment was done at least thrice, and representative data are shown. Data in the bar graph are given as the means ± SE. The means were checked for statistical difference with the use of a t test, and P values <0.05 were considered significant (*, P < 0.05; **, P < 0.01; ***, P < 0.001).

EPOX inhibits angiogenesis in vitro and in vivo

To test the hypothesis that EPOX may inhibit angiogenesis, initially we determined the effect of EPOX with the Matrigel plug assay. The Matrigel plugs in the control groups were visually bloodier than the EPOX-treated ones, suggesting a higher level of angiogenesis (Fig. 1A, top). Histologic analysis of the Matrigel pellets identified more erythrocyte-containing vessels within the control than in the EPOX-treated (Fig. 1A, middle). Quantification of angiogenesis by hemoglobin content further showed that EPOX inhibited angiogenic response in a dose-dependent manner (Fig. 1A, bottom), suggesting that EPOX attenuated angiogenesis in vivo. We proceeded to examine whether EPOX inhibits tube formation, which is a well-accepted technique to measure in vitro angiogenesis. Treatment with EPOX significantly reduced this process (Fig. 1B).

Fig. 1.

EPOX inhibits angiogenesis. A, antiangiogenesis effect of EPOX in in vivo Matrigel plug assay. Top, macroscopic appearance of Matrigel plugs after harvest; middle, Masson's trichrome staining of cross-sections from Matrigel plugs; bottom, quantitation of active vasculature inside the Matrigel by measurement of hemoglobin content. Hemoglobin was quantified and presented compared with the control. B, tubular morphogenesis was blocked by EPOX. Tube length was quantified by measuring the length of tubes in three randomly chosen fields from each well with the use of Image-Pro Plus software and was calculated against DMSO control. C, viability measurements. Cells were treated with the indicated concentrations of EPOX for 24 or 48 h. Cell number was determined by the MTT reduction assay, and viability is expressed as the percentage of living cells present in vehicle-treated preparations. D, time-dependent changes in the sub-G1 population after EPOX treatment. Cells were incubated with 3 μmol/L EPOX and analyzed for propidium iodide–stained DNA content by flow cytometry. Values, percentage of cells with hypodiploid DNA content. MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide.

Fig. 1.

EPOX inhibits angiogenesis. A, antiangiogenesis effect of EPOX in in vivo Matrigel plug assay. Top, macroscopic appearance of Matrigel plugs after harvest; middle, Masson's trichrome staining of cross-sections from Matrigel plugs; bottom, quantitation of active vasculature inside the Matrigel by measurement of hemoglobin content. Hemoglobin was quantified and presented compared with the control. B, tubular morphogenesis was blocked by EPOX. Tube length was quantified by measuring the length of tubes in three randomly chosen fields from each well with the use of Image-Pro Plus software and was calculated against DMSO control. C, viability measurements. Cells were treated with the indicated concentrations of EPOX for 24 or 48 h. Cell number was determined by the MTT reduction assay, and viability is expressed as the percentage of living cells present in vehicle-treated preparations. D, time-dependent changes in the sub-G1 population after EPOX treatment. Cells were incubated with 3 μmol/L EPOX and analyzed for propidium iodide–stained DNA content by flow cytometry. Values, percentage of cells with hypodiploid DNA content. MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide.

Close modal

In an attempt to discover the mechanism of EPOX to inhibit angiogenesis, we first did MTT and bromodeoxyuridine incorporation assays to measure the effect of EPOX on the growth of HUVECs. EPOX was found to decrease cell viability (Fig. 1C) and DNA synthesis (Supplementary Fig. S1C) in a concentration-dependent manner. The EAhy926 cell line has been suggested to be an alternative model for HUVECs to study the control of pathologic angiogenesis by endogenous factors and pharmacologic compounds (23). Consistently, EPOX inhibited EAhy926 cell growth (Supplementary Fig. S2A) and tube formation (Supplementary Fig. S2B).

To examine whether EPOX-induced growth inhibition was associated with cell cycle regulation, the cell cycle distributions of EPOX-treated cells were analyzed by flow cytometry. As illustrated in Fig. 1D, EPOX treatment increased the percentage of cells in the sub-G1 phase. EPOX-treated HUVECs exhibited both rounding and blebbing during morphologic examination (Supplementary Fig. S2C, top). To verify the occurrence of apoptosis, cells were inspected after DAPI staining. In the absence of EPOX, cells exhibited nuclei with homogeneous chromatin distribution. However, EPOX treatment induced nuclear fragmentation and chromatin condensation (Supplementary Fig. S2C, bottom). DNA fragmentation was also increased in EPOX-treated cells in a concentration-dependent manner (Supplementary Fig. S2D). Taken together, the results suggest that EPOX treatment induces apoptosis of endothelial cells.

EPOX treatment activates the mitochondria-mediated apoptosis pathway

The activation of selected caspases serves to identify the involvement of specific apoptotic signaling pathways. To further characterize the mechanism through which EPOX triggers apoptosis, the activation states of caspase-3, caspase-8, and caspase-9 were therefore examined. Notably, EPOX treatment resulted in the cleavage and activation of caspase-9 and caspase-3 in HUVECs (Fig. 2A).

Fig. 2.

EPOX promotes activation of the mitochondrial pathway of apoptosis. A, caspase activation. Cells were treated with EPOX (3 μmol/L) for the indicated times before immunoblotting and detection of caspase activity. Arrowhead, cleaved forms of caspases. Means ± SE of three independent determination of caspase activity were checked for statistical difference. *, P < 0.05; **, P < 0.01; ***, P < 0.001, compared with nontreated cells. B, mitochondrial membrane potential. Cells were treated with EPOX (3 μmol/L) for the indicated times and then harvested for the determination of mitochondrial membrane potential through measuring rhodamine 123 fluorescence intensity by flow cytometry. Results are compared with the DMSO control. C, translocation of cytochrome c. Translocation of cytochrome c from the mitochondria to the cytosol after 4 or 6 h of treatment without or with EPOX (3 μmol/L) was measured by subcellular fractionation and immunoblotting.

Fig. 2.

EPOX promotes activation of the mitochondrial pathway of apoptosis. A, caspase activation. Cells were treated with EPOX (3 μmol/L) for the indicated times before immunoblotting and detection of caspase activity. Arrowhead, cleaved forms of caspases. Means ± SE of three independent determination of caspase activity were checked for statistical difference. *, P < 0.05; **, P < 0.01; ***, P < 0.001, compared with nontreated cells. B, mitochondrial membrane potential. Cells were treated with EPOX (3 μmol/L) for the indicated times and then harvested for the determination of mitochondrial membrane potential through measuring rhodamine 123 fluorescence intensity by flow cytometry. Results are compared with the DMSO control. C, translocation of cytochrome c. Translocation of cytochrome c from the mitochondria to the cytosol after 4 or 6 h of treatment without or with EPOX (3 μmol/L) was measured by subcellular fractionation and immunoblotting.

Close modal

Caspase-9 has been proposed as the predominant initiator caspase in the mitochondria-dependent intrinsic apoptosis pathway. In response to apoptotic stimuli, dissipation of the mitochondrial membrane potential results in the release of cytochrome c from the mitochondrial intermembrane space. Upon release to the cytosol, cytochrome c binds Apaf-1, and then recruits and activates procaspase-9. The mitochondrial membrane potential and release of cytochrome c from the mitochondria to the cytosol were therefore measured in untreated and EPOX-treated HUVECs. Treatment with EPOX decreased the mitochondrial membrane potential in a time-dependent manner (Fig. 2B). As shown in Fig. 2C, the efflux of cytochrome c from mitochondria was detected in EPOX-treated cells simultaneously with the accumulation of cytochrome c in the cytosol. Taken together, these findings are consistent with the hypothesis that EPOX induces apoptosis in HUVECs through a mitochondria-mediated pathway.

EPOX treatment induces rapid proteasome-dependent degradation of Mcl-1

Bcl-2 proteins, a family that includes both proapoptotic and antiapoptotic members, are critical regulators of the mitochondrial pathway of apoptosis. Proapoptotic members, such as Bax and Bak, promote the release of cytochrome c from the mitochondria, whereas antiapoptotic members, such as Bcl-2, Bcl-xL, and Mcl-1, prevent release. The balance of proapoptotic and antiapoptotic Bcl-2 proteins therefore decides the fate of the cell. Because the above findings reveal that EPOX activates the mitochondrial pathway of cell apoptosis, it was of interest to identify the Bcl-2 family members involved in the apoptosis triggered by EPOX. EPOX treatment induced the rapid down-regulation of Mcl-1, whereas no significant changes in expression of Bcl-2, Bcl-xL, Bax, Bak, or Bim were observed (Fig. 3A). If loss of Mcl-1 after EPOX treatment was responsible for the subsequent induction of apoptosis, then maintenance of Mcl-1 should prevent apoptosis. As shown in Fig. 3B, transient transfection of Mcl-1 partially overcomes the cleavages of caspase-3 and poly(ADP-ribose) polymerase, and the loss of viability in response to EPOX treatment. These observations support the hypothesis that down-regulation of Mcl-1 contributes to EPOX-induced apoptosis in HUVECs.

Fig. 3.

EPOX induces proteasome-dependent degradation of Mcl-1. A, Mcl-1, but not other Bcl-2 family members, is down-regulated during treatment with EPOX. Cells were exposed to EPOX (3 μmol/L) for the indicated times. Extracts were then subjected to western blotting with the use of antibodies to various Bcl-2 family members. B, Mcl-1 overexpression attenuates the effects of EPOX. Cells were transfected with vector or HA-Mcl-1 at 48 h before addition of vehicle or EPOX (3 μmol/L). Top, results of immunoblotting after 6 h of treatments; bottom, cell viabilities after 24 h of treatments. C, ubiquitination and proteasomal degradation of Mcl-1 in EPOX-treated cells. Top, degradation. Cells were treated without or with the proteasome inhibitor MG132 (10 μmol/L) for the indicated times in the presence of EPOX. Cell extracts were then prepared followed by immunoblotting. Bottom, ubiquitination. Cells transfected with HA-ubiquitin were treated for 4 h without or with EPOX (3 μmol/L) in the presence of MG132 (10 μmol/L). Lysates were subjected to immunoprecipitation with anti–Mcl-1 antibody. D, PARP processing and cell death in response to EPOX are reduced by caspase inhibition. Preparations were pretreated without or with z-VAD-fmk (ZVAD; 100 μmol/L) for 1 h. Top, results of immunoblotting after 6 h of EPOX treatments; bottom, results of MTT reduction assays done after 24 h of incubation without or with EPOX (3 μmol/L). PARP, poly(ADP-ribose) polymerase.

Fig. 3.

EPOX induces proteasome-dependent degradation of Mcl-1. A, Mcl-1, but not other Bcl-2 family members, is down-regulated during treatment with EPOX. Cells were exposed to EPOX (3 μmol/L) for the indicated times. Extracts were then subjected to western blotting with the use of antibodies to various Bcl-2 family members. B, Mcl-1 overexpression attenuates the effects of EPOX. Cells were transfected with vector or HA-Mcl-1 at 48 h before addition of vehicle or EPOX (3 μmol/L). Top, results of immunoblotting after 6 h of treatments; bottom, cell viabilities after 24 h of treatments. C, ubiquitination and proteasomal degradation of Mcl-1 in EPOX-treated cells. Top, degradation. Cells were treated without or with the proteasome inhibitor MG132 (10 μmol/L) for the indicated times in the presence of EPOX. Cell extracts were then prepared followed by immunoblotting. Bottom, ubiquitination. Cells transfected with HA-ubiquitin were treated for 4 h without or with EPOX (3 μmol/L) in the presence of MG132 (10 μmol/L). Lysates were subjected to immunoprecipitation with anti–Mcl-1 antibody. D, PARP processing and cell death in response to EPOX are reduced by caspase inhibition. Preparations were pretreated without or with z-VAD-fmk (ZVAD; 100 μmol/L) for 1 h. Top, results of immunoblotting after 6 h of EPOX treatments; bottom, results of MTT reduction assays done after 24 h of incubation without or with EPOX (3 μmol/L). PARP, poly(ADP-ribose) polymerase.

Close modal

To determine how EPOX reduces Mcl-1 expression, we first measured Mcl-1 mRNA content by reverse transcription-PCR and found that Mcl-1 mRNA values were unaffected by EPOX (Supplementary Fig. S3A). We then investigated the degradation of Mcl-1. As shown in Fig. 3C, Mcl-1 degradation in EPOX-treated HUVECs was almost fully blocked by exposure to the proteasomal inhibitor MG132. An increase in ubiquitination of Mcl-1 was also observed for EPOX-treated compared with nontreated preparations. Consistently, in the presence of cycloheximide, which inhibits de novo protein synthesis, the rate of degradation of Mcl-1 was significantly greater in EPOX-treated compared with vehicle-treated cells (Supplementary Fig. S3B). These findings support the proposal that EPOX decreases Mcl-1 level through activation of the ubiquitin-dependent and proteasome-dependent pathways.

Mcl-1 is also described as a substrate for caspase-mediated degradation. In previous data, cleavage of caspase-3 was observed in EPOX-treated HUVECs. To ascertain whether caspase activation contributes to Mcl-1 down-regulation in these cells, experiments were also conducted in the presence of ZVAD-fmk, an established caspase inhibitor. In EPOX-treated HUVECs exposed to ZVAD-fmk, caspase-3 and poly(ADP-ribose) polymerase processing were significantly reduced, and viability was increased compared with preparations not treated with ZVAD-fmk (Fig. 3D). However, exposure to ZVAD-fmk did not notably influence Mcl-1 down-regulation in these cells, suggesting that this down-regulation occurred upstream of caspase activation.

Mcl-1–free Bim generated in EPOX-treated HUVECs activates Bax

Mcl-1 is known to sequester Bim in an inactive state. Experiments were therefore done to ascertain whether Mcl-1/Bim complexes are affected in EPOX-treated HUVECs. Endogenous Mcl-1/Bim complexes were detectable in extracts of nontreated HUVECs. After 6 hours of EPOX treatment, however, an increase in Mcl-1–free Bim was observed (Fig. 4A). To examine the effects of Bim on the apoptotic response to EPOX, Bim was knocked down by RNA interference (RNAi). Knockdown of Bim conferred partial resistance to EPOX-induced caspase-3 and poly(ADP-ribose) polymerase processing, and the subsequent apoptosis (Fig. 4B).

Fig. 4.

Mcl-1–free Bim generated by EPOX treatment activates Bax in endothelial cells. A, dissociation of Bim from Mcl-1 in EPOX-treated cells. Cells were incubated without or with EPOX (3 μmol/L) for 6 h. Lysates were treated with anti–Mcl-1 antibody. Bound (IP pellet), unbound fractions (deplete supernatant, 5% of treated lysates), and total cell lysate were subjected to western blotting. B, knockdown of Bim by Bim siRNA attenuates EPOX-induced apoptosis. Cells were pretreated with scramble or Bim siRNA for 72 h before incubations without or with EPOX (3 μmol/L). Top, results of immunoblotting after 6 h of incubation; bottom, viabilities after 24 h of incubation. C, Bax translocates from the cytosol to the mitochondria after treatment with EPOX. Cells were incubated without or with EPOX (3 μmol/L) for 4 or 6 h. Cytosolic and mitochondrial fractions were prepared and examined by western blotting. D, confocal microscopy revealing the translocation of Bax from cytosol to mitochondria in response to EPOX treatment. Cells were treated without or with EPOX (3 μmol/L) for 6 h and stained with anti-Bax antibodies followed by FITC-conjugated secondary antibody. Mitochondria were stained with MitoTracker.

Fig. 4.

Mcl-1–free Bim generated by EPOX treatment activates Bax in endothelial cells. A, dissociation of Bim from Mcl-1 in EPOX-treated cells. Cells were incubated without or with EPOX (3 μmol/L) for 6 h. Lysates were treated with anti–Mcl-1 antibody. Bound (IP pellet), unbound fractions (deplete supernatant, 5% of treated lysates), and total cell lysate were subjected to western blotting. B, knockdown of Bim by Bim siRNA attenuates EPOX-induced apoptosis. Cells were pretreated with scramble or Bim siRNA for 72 h before incubations without or with EPOX (3 μmol/L). Top, results of immunoblotting after 6 h of incubation; bottom, viabilities after 24 h of incubation. C, Bax translocates from the cytosol to the mitochondria after treatment with EPOX. Cells were incubated without or with EPOX (3 μmol/L) for 4 or 6 h. Cytosolic and mitochondrial fractions were prepared and examined by western blotting. D, confocal microscopy revealing the translocation of Bax from cytosol to mitochondria in response to EPOX treatment. Cells were treated without or with EPOX (3 μmol/L) for 6 h and stained with anti-Bax antibodies followed by FITC-conjugated secondary antibody. Mitochondria were stained with MitoTracker.

Close modal

Once liberated from Mcl-1, Bim is able to activate Bax directly (24). A change in Bax conformation to expose its N-terminal and C-terminal regions is intimately associated with the mitochondrial targeting and proapoptotic activity of the protein (25). To ascertain whether Bax is activated in EPOX-treated HUVECs, the protein was immunoprecipitated from cell extracts with the use of an antibody (anti-Bax 6A7) that recognizes only the conformationally altered protein. As shown in Fig. 4B, Bax underwent a conformational change in EPOX-treated cells. However, knockdown of Bim attenuated Bax activation in EPOX-treated cells, implying that Bim is involved in the activation. Subcellular fractionation (Fig. 4C) and confocal microscopy (Fig. 4D) confirmed Bax translocation to mitochondria during EPOX treatment. In untreated cells, the cytoplasm displayed diffuse immunostaining for Bax, whereas a punctate distribution, colocalizing with MitoTracker staining, was observed after EPOX treatment.

Glutathione depletion is associated with mitochondrial alterations in response to EPOX

To further characterize the upstream signals involved in the induction of apoptosis in EPOX-treated HUVECs, specific pharmacologic agents were tested for their capacity to ameliorate the apoptosis. Modulators of the “oxidant,” “calcium,” and “arachidonic acid” pathways associated with the induction of endothelial cell apoptosis in response to stimuli, including lipopolysaccharide, hypoxia, hydrogen peroxide, tumor necrosis factor α, IFN γ, activated neutrophils, or oxidized low-density lipoprotein (26), were tested. As shown in Fig. 5A, only glutathione ethyl ester, a cell-permeable analogue of GSH, significantly reduced EPOX-induced cell death.

Fig. 5.

Glutathione depletion is responsible for mitochondrial alterations produced by EPOX treatment. A, effect of various pharmacologic agents on EPOX-induced HUVEC cytotoxicity. Pharmacologic inhibitors, documented to protect against oxidant-mediated, calcium-mediated, and arachidonic acid pathway–mediated endothelial cell death, were preincubated with HUVECs and then incubated with 3 μmol/L EPOX for 24 h. Cell number was determined by MTT assay and was calculated as the percentage of vehicle treatment. B, GSHee increases viability of EPOX-treated cells. Cells were pretreated with GSHee at the indicated concentrations before incubations with EPOX (3 μmol/L). Viability was determined after 24 h through MTT assay. C, cellular GSH content declines during treatment with EPOX. Cells were treated with EPOX (3 μmol/L) for the indicated times. After treatment, cells were loaded with monochlorobimane (100 μmol/L) followed by measurements of fluorescence intensity with a microplate reading fluorometer. D, GSHee reduces the apoptosis-associated events in EPOX-treated cells. Cells were pretreated without or with GSHee (3 mmol/L) and then incubated for 6 h with EPOX (3 μmol/L). Lysates were subjected to immunoblotting. GSHee, glutathione ethyl ester.

Fig. 5.

Glutathione depletion is responsible for mitochondrial alterations produced by EPOX treatment. A, effect of various pharmacologic agents on EPOX-induced HUVEC cytotoxicity. Pharmacologic inhibitors, documented to protect against oxidant-mediated, calcium-mediated, and arachidonic acid pathway–mediated endothelial cell death, were preincubated with HUVECs and then incubated with 3 μmol/L EPOX for 24 h. Cell number was determined by MTT assay and was calculated as the percentage of vehicle treatment. B, GSHee increases viability of EPOX-treated cells. Cells were pretreated with GSHee at the indicated concentrations before incubations with EPOX (3 μmol/L). Viability was determined after 24 h through MTT assay. C, cellular GSH content declines during treatment with EPOX. Cells were treated with EPOX (3 μmol/L) for the indicated times. After treatment, cells were loaded with monochlorobimane (100 μmol/L) followed by measurements of fluorescence intensity with a microplate reading fluorometer. D, GSHee reduces the apoptosis-associated events in EPOX-treated cells. Cells were pretreated without or with GSHee (3 mmol/L) and then incubated for 6 h with EPOX (3 μmol/L). Lysates were subjected to immunoblotting. GSHee, glutathione ethyl ester.

Close modal

Supplementation with glutathione ethyl ester was done for 6 hours followed by replacement with nonsupplemented medium to avoid nonspecific reactions between glutathione ethyl ester and EPOX. Supplementation was found to reverse the effect of EPOX on cell growth (Fig. 5B) and tube formation (Supplementary Fig. S3C). With regard to the importance of glutathione for the susceptibility of endothelial cells to EPOX, it was of interest to observe the changes in intracellular GSH levels after EPOX treatment. Cells were incubated with monochlorobimane, a well-established GSH-specific probe, and then analyzed for GSH content with a fluorescence microplate reader. EPOX treatment induced the depletion of GSH in a time-dependent manner (Fig. 5C). When fluorescence microscopy was used, consistently EPOX treatment was found to cause a striking decrease in monochlorobimane fluorescence (Supplementary Fig. S3D). These findings implicate GSH depletion in the induction of apoptosis by EPOX.

Reduced GSH levels are frequently associated with activation of apoptosis through mitochondrial pathways (27). Studies were therefore done to explore the role of GSH in the mitochondrial alterations observed in response to EPOX treatment. As shown in Supplementary Fig. S4A, glutathione ethyl ester supplementation significantly restored the collapse of mitochondria membrane potential in response to EPOX treatment. Additionally, pretreatment with glutathione ethyl ester reduced the down-regulation of Mcl-1, the conformational change in Bax, and the cleavages of caspase-3 and poly(ADP-ribose) polymerase after EPOX treatment (Fig. 5D). Depletion of GSH therefore seems to serve as an upstream event for induction of Mcl-1 degradation by EPOX.

Down-regulation of Mcl-1 by EPOX involves inhibition of ERK activation

The stability of Mcl-1 changes in response to phosphorylation by ERK, JNK, and GSK-3β (16, 28, 29).Among these three kinases, ERK was found to be the most significantly inactivated by EPOX treatment (Fig. 6A). When a constitutively active MEK that activated ERK was transiently transfected into HUVECs, the ERK inactivation by EPOX was partially restored. Interestingly, Mcl-1 down-regulation, downstream apoptotic pathway activation (Fig. 6B), and the loss of viability (Supplementary Fig. S4B) in response to EPOX were also attenuated, consistent with a role for ERK inactivation in EPOX-induced Mcl-1 degradation.

Fig. 6.

Inhibition of ERK activation contributed to the down-regulation of Mcl-1 in EPOX-treated cells. A, ERK, but not JNK or GSK-3β, is inactivated by EPOX, and GSHee reduces the inactivation. Cells were pretreated without or with GSHee (3 mmol/L) before 6-h incubations with EPOX (3 μmol/L). Lysates were subjected to immunoblotting. B, MEK overexpression reduces the apoptosis-associated events in EPOX-treated cells. Cells were transfected with vector or constitutively active MEK at 48 h before incubations for 6 h with EPOX (3 μmol/L). Lysates were subjected to immunoblotting for the indicated proteins. C, overexpression of constitutively active MEK reduces dephosphorylation and ubiquitination of Mcl-1 in EPOX-treated cells. Cells were either cotransfected with HA-ubiquitin and MEK, or HA-ubiquitin only. Incubations were then conducted for 4 h with MG132 (10 μmol/L) in the absence or presence of EPOX (3 μmol/L) as indicated. D, GSHee supplementation attenuates the effect of EPOX on Mcl-1. Cells transfected with HA-ubiquitin were pretreated without or with GSHee (3 mmol/L) and incubated for 4 h with MG132 (10 μmol/L) in the absence or presence of EPOX (3 μmol/L) as indicated. JNK, c-Jun-NH2-kinase.

Fig. 6.

Inhibition of ERK activation contributed to the down-regulation of Mcl-1 in EPOX-treated cells. A, ERK, but not JNK or GSK-3β, is inactivated by EPOX, and GSHee reduces the inactivation. Cells were pretreated without or with GSHee (3 mmol/L) before 6-h incubations with EPOX (3 μmol/L). Lysates were subjected to immunoblotting. B, MEK overexpression reduces the apoptosis-associated events in EPOX-treated cells. Cells were transfected with vector or constitutively active MEK at 48 h before incubations for 6 h with EPOX (3 μmol/L). Lysates were subjected to immunoblotting for the indicated proteins. C, overexpression of constitutively active MEK reduces dephosphorylation and ubiquitination of Mcl-1 in EPOX-treated cells. Cells were either cotransfected with HA-ubiquitin and MEK, or HA-ubiquitin only. Incubations were then conducted for 4 h with MG132 (10 μmol/L) in the absence or presence of EPOX (3 μmol/L) as indicated. D, GSHee supplementation attenuates the effect of EPOX on Mcl-1. Cells transfected with HA-ubiquitin were pretreated without or with GSHee (3 mmol/L) and incubated for 4 h with MG132 (10 μmol/L) in the absence or presence of EPOX (3 μmol/L) as indicated. JNK, c-Jun-NH2-kinase.

Close modal

Phosphorylation of Mcl-1 at T163, the conserved ERK site located within the proline, glutamic acid, serine, and threonine (PEST) region (16), was abolished by treatments with the MEK inhibitors PD98059 and U0126 (Supplementary Fig. S4C). Domina et al. (28) showed that phosphorylation of Mcl-1 at T163 by ERK serves to prolong the half-life of Mcl-1. Half-life prolongation is attributable to the binding of Pin1 at the phosphorylated T163 site, which serves to protect Mcl-1 from degradation by interfering with its ubiquitination (29, 30). Accordingly, treatments with MEK inhibitors promoted Mcl-1 ubiquitination (Supplementary Fig. S4C).

Experiments were done to examine T163 phosphorylation of Mcl-1 after EPOX treatment and to ascertain whether ERK activation attenuates the effect of EPOX. As shown in Fig. 6C, overexpression of constitutively active MEK, which served to activate ERK, reduced the T163 dephosphorylation and ubiquitination of Mcl-1 in response to EPOX treatment. Interestingly, glutathione ethyl ester supplementation was observed to reverse the effects of EPOX on ERK inactivation, on Mcl-1 dephosphorylation, and on Mcl-1 ubiquitination and degradation (Figs. 5D, and 6A and D). These observations, which are consistent with those of others (28, 31), highlight the involvement of the ERK in the regulation of Mcl-1 stability. These findings also reveal that ERK is involved in the down-regulation of Mcl-1 in EPOX-treated HUVECs.

The increasing use of antiangiogenic drugs for the treatment of cancer is the consequence of decades of extensive basic and clinical research. However, the clinical benefits of antiangiogenic therapy are relatively modest, with survival measured in months (32). Better understanding of the molecular mechanisms governing angiogenesis and of the molecular responses to antiangiogenic therapies are required for therapeutic improvements. The present study reveals that the xanthone derivative EPOX induces apoptosis in endothelial cells through pathways initiated by GSH depletion and Mcl-1 degradation. In this regard, EPOX was also examined for apoptotic effects with the use of a panel of normal cells and cancer cell lines (Supplementary Table S1). This agent produced prominent apoptotic effects in several types of cancer cells, and modest apoptotic effects in normal prostatic cells and fibroblasts. In a colon cancer xenograft model, treatment with EPOX caused a significant inhibition of tumor growth (Supplementary Fig. S5A) without overt toxicity (Supplementary Fig. S5B). We also observed that the EPOX-treated tumor had less CD31+ vessel structures than the control group (Supplementary Fig. S5C), supporting the potential of EPOX as an angiognenesis inhibitor for cancer therapy.

GSH effectively scavenges free radicals and other reactive oxygen species, both directly and indirectly through enzymatic reactions. Depletion of intracellular GSH content is frequently accompanied by a concomitant increase in reactive oxygen species. However, GSH depletion could lower the reducing capacity of the cells, thereby promoting oxidative stress independently of reactive oxygen species (33). In the present study, supplementation with glutathione ethyl ester, but not with other antioxidants, ameliorated EPOX-induced apoptosis. Furthermore, EPOX treatment did not promote accumulation of reactive oxygen species as monitored through use of 2′,7′-dichlorodihydrofluorecein diacetate or hydroxyethidine (data not shown). Reactive oxygen species have been shown to activate ERK in a variety of cell types (34), although the mechanisms responsible for the activation have not been identified clearly. In the current study, EPOX treatment was observed to suppress activation of ERK in endothelial cells. Taken together, these findings support the hypothesis that EPOX induces endothelial cell apoptosis independently of reactive oxygen species. Further studies are clearly required to determine the mechanism whereby GSH depletion leads to ERK inactivation after EPOX treatment.

Because most tumors are recognized to have defects in the p53 pathway or to overexpress a Bcl-2 homologue, the therapeutic potential of anticancer drugs that, like the BH3 domain, bind one or more Bcl-2 homologues and trigger apoptosis are of significant current interest. Such“BH3 mimetics” should prove more effective than antitumor agents acting far upstream (35). A structure-based approach recently resulted in the development of ABT-737, an agent that binds strongly to Bcl-2, Bcl-xL, and Bcl-w, but not to Mcl-1 (36). Because inactivation of Mcl-1 is critical for cell death to proceed (37), knockdown of Mcl-1 with the use of siRNA was found to sensitize cells to the effects of ABT-737 (38). Small molecules that target Mcl-1, such as obatoclax (39), should serve to complement the antitumor actions of ABT-737. However, it should be noted that the rapid and marked down-regulation of Mcl-1 through proteasome-dependent degradation, as observed in the present study, was found to contribute to EPOX-induced apoptosis. EPOX in combination with ABT-737 should therefore represent an effective therapy for induction of tumor cell apoptosis (40).

Current evidence supports the importance of Raf/MEK/ERK signaling in stimulation of angiogenesis. Raf-1 knockout mice die from vascular defects in the yolk sac in association with an increased propensity for apoptosis (40). In response to intratumor hypoxia, the proangiogenic factors vascular endothelial growth factor, basic fibroblast growth factor, and platelet-derived growth factor are up-regulated and secreted. Interestingly, all of these growth factors exert their signaling effects through the Raf/MEK/ERK pathway. Furthermore, sorafenib, a Raf kinase inhibitor, strongly inhibits angiogenesis in tumor xenograft models (41). Although sorafenib was recently approved by the Food and Drug Administration for treating advanced renal cell carcinoma (42), the molecular mechanism through which this agent promotes apoptosis is not completely characterized. However, sorafenib was recently reported to enhance proteasome-mediated Mcl-1 degradation (31) and to increase apoptosis induced by tumor necrosis factor–related apoptosis-inducing ligand through down-regulation of Mcl-1 (43). The findings of the present study highlight the involvement of the ERK in the regulation of Mcl-1 stability. Recent findings in our laboratory revealed that ERK could phosphorylate Mcl-1 at Thr 92 and Thr 163, which is required for the association of Mcl-1 and Pin1 (30). A novel mechanism was identified linking the ERK-Pin1 pathway with Mcl-1–mediated chemoresistance. Consistent with these findings, the inhibition of ERK activation in response to EPOX treatment was also found to promote the dissociation of Mcl-1 from Pin1.5

5Unpublished data.

Supplementary Fig. S6 presents a model, based on the findings of the present report, for EPOX-induced apoptosis in endothelial cells. In response to EPOX treatment, the depletion of GSH signals ERK inactivation, which results in the dephosphorylation and down-regulation of Mcl-1 through the proteasome-mediated degradation. The liberation of Bim from Mcl-1 serves to induce the conformational change in Bax required for its activation. Upon translocation to the mitochondria, the activated Bax triggers apoptosis through the release of cytochrome c, and activations of caspase-9 and caspase-3. It is therefore proposed that therapies designed to manipulate GSH content, the ERK/Mcl-1 pathway, and crosstalk among Bcl-2 members could prove effective for suppression of angiogenesis.

No potential conflicts of interest were disclosed.

We thank Hsin-Fang Yang-Yen and Pier Paolo Pandolfi for providing reagents; Kuo-Shyan Lu for the EAhy926 cells; Chieh-Yu Peng, Ying-Cheng Chen, Yao-Ting Huang, Dong-Ming Huang, Dung-Fang Lee, Chun-Te Chen, Jung-Mao Hsu, and Jer-Yen Yang for technical support; and Chin-Yu Lai, Anita Shin-Yuan Chang, Margaret A. Brostrom, Jeng C. Cheng, and Stephanie A. Miller for editing the manuscript.

1
Folkman
J
. 
Role of angiogenesis in tumor growth and metastasis
.
Semin Oncol
2002
;
29
:
15
8
.
2
Ou-Yang
F
,
Lan
KL
,
Chen
CT
, et al
. 
Endostatin-cytosine deaminase fusion protein suppresses tumor growth by targeting neovascular endothelial cells
.
Cancer Res
2006
;
66
:
378
84
.
3
Duval
H
,
Harris
M
,
Li
J
,
Johnson
N
,
Print
C
. 
New insights into the function and regulation of endothelial cell apoptosis
.
Angiogenesis
2003
;
6
:
171
83
.
4
Dimmeler
S
,
Zeiher
AM
. 
Endothelial cell apoptosis in angiogenesis and vessel regression
.
Circ Res
2000
;
87
:
434
9
.
5
Browder
T
,
Butterfield
CE
,
Kraling
BM
, et al
. 
Antiangiogenic scheduling of chemotherapy improves efficacy against experimental drug-resistant cancer
.
Cancer Res
2000
;
60
:
1878
86
.
6
Kerbel
RS
,
Kamen
BA
. 
The anti-angiogenic basis of metronomic chemotherapy
.
Nat Rev Cancer
2004
;
4
:
423
36
.
7
Folkman
J
. 
Angiogenesis and apoptosis
.
Semin Cancer Biol
2003
;
13
:
159
67
.
8
Okada
H
,
Mak
TW
. 
Pathways of apoptotic and non-apoptotic death in tumour cells
.
Nat Rev Cancer
2004
;
4
:
592
603
.
9
Moran
LK
,
Gutteridge
JM
,
Quinlan
GJ
. 
Thiols in cellular redox signalling and control
.
Curr Med Chem
2001
;
8
:
763
72
.
10
McEligot
AJ
,
Yang
S
,
Meyskens
FL
. 
Redox regulation by intrinsic species and extrinsic nutrients in normal and cancer cells
.
Annu Rev Nutr
2005
;
25
:
261
95
.
11
Pinto
MM
,
Sousa
ME
,
Nascimento
MS
. 
Xanthone derivatives: new insights in biological activities
.
Curr Med Chem
2005
;
12
:
2517
38
.
12
McKeage
MJ
,
Von
PJ
,
Reck
M
, et al
. 
Randomised phase II study of ASA404 combined with carboplatin and paclitaxel in previously untreated advanced non-small cell lung cancer
.
Br J Cancer
2008
;
99
:
2006
12
.
13
Baguley
BC
. 
Antivascular therapy of cancer: DMXAA
.
Lancet Oncol
2003
;
4
:
141
8
.
14
Lin
CN
,
Liou
SJ
,
Lee
TH
,
Chuang
YC
,
Won
SJ
. 
Xanthone derivatives as potential anti-cancer drugs
.
J Pharm Pharmacol
1996
;
48
:
539
44
.
15
Liu
HS
,
Lin
CN
,
Won
SJ
. 
Antitumor effect of 2,6-di(2,3-epoxypropoxy)xanthone on tumor cell line
.
Anticancer Res
1997
;
17
:
1107
14
.
16
Ding
Q
,
He
X
,
Hsu
JM
, et al
. 
Degradation of Mcl-1 by β-TrCP mediates glycogen synthase kinase 3-induced tumor suppression and chemosensitization
.
Mol Cell Biol
2007
;
27
:
4006
17
.
17
Peng
CY
,
Pan
SL
,
Lee
KH
,
Bastow
KF
,
Teng
CM
. 
Molecular mechanism of the inhibitory effect of KS-5 on bFGF-induced angiogenesis in vitro and in vivo
.
Cancer Lett
2008
;
263
:
114
21
.
18
Dong
LF
,
Swettenham
E
,
Eliasson
J
, et al
. 
Vitamin E analogues inhibit angiogenesis by selective induction of apoptosis in proliferating endothelial cells: the role of oxidative stress
.
Cancer Res
2007
;
67
:
11906
13
.
19
Huang
YT
,
Huang
DM
,
Guh
JH
,
Chen
IL
,
Tzeng
CC
,
Teng
CM
. 
CIL-102 interacts with microtubule polymerization and causes mitotic arrest following apoptosis in the human prostate cancer PC-3 cell line
.
J Biol Chem
2005
;
280
:
2771
9
.
20
Chen
WY
,
Wu
CC
,
Lan
YH
,
Chang
FR
,
Teng
CM
,
Wu
YC
. 
Goniothalamin induces cell cycle-specific apoptosis by modulating the redox status in MDA-MB-231 cells
.
Eur J Pharmacol
2005
;
522
:
20
9
.
21
Sun
HL
,
Liu
YN
,
Huang
YT
, et al
. 
YC-1 inhibits HIF-1 expression in prostate cancer cells: contribution of Akt/NF-κB signaling to HIF-1α accumulation during hypoxia
.
Oncogene
2007
;
26
:
3941
51
.
22
Yamaguchi
H
,
Paranawithana
SR
,
Lee
MW
,
Huang
Z
,
Bhalla
KN
,
Wang
HG
. 
Epothilone B analogue (BMS-247550)-mediated cytotoxicity through induction of Bax conformational change in human breast cancer cells
.
Cancer Res
2002
;
62
:
466
71
.
23
Shtivelband
MI
,
Juneja
HS
,
Lee
S
,
Wu
KK
. 
Aspirin and salicylate inhibit colon cancer medium- and VEGF-induced endothelial tube formation: correlation with suppression of cyclooxygenase-2 expression
.
J Thromb Haemost
2003
;
1
:
2225
33
.
24
Kuwana
T
,
Bouchier-Hayes
L
,
Chipuk
JE
, et al
. 
BH3 domains of BH3-only proteins differentially regulate Bax-mediated mitochondrial membrane permeabilization both directly and indirectly
.
Mol Cell
2005
;
17
:
525
35
.
25
Wolter
KG
,
Hsu
YT
,
Smith
CL
,
Nechushtan
A
,
Xi
XG
,
Youle
RJ
. 
Movement of Bax from the cytosol to mitochondria during apoptosis
.
J Cell Biol
1997
;
139
:
1281
92
.
26
Osband
AJ
,
Deitch
EA
,
Lu
Q
, et al
. 
The role of oxidant-mediated pathways in the cytotoxicity of endothelial cells exposed to mesenteric lymph from rats subjected to trauma-hemorrhagic shock
.
Shock
2003
;
20
:
269
73
.
27
Hammond
CL
,
Lee
TK
,
Ballatori
N
. 
Novel roles for glutathione in gene expression, cell death, and membrane transport of organic solutes
.
J Hepatol
2001
;
34
:
946
54
.
28
Domina
AM
,
Vrana
JA
,
Gregory
MA
,
Hann
SR
,
Craig
RW
. 
MCL1 is phosphorylated in the PEST region and stabilized upon ERK activation in viable cells, and at additional sites with cytotoxic okadaic acid or taxol
.
Oncogene
2004
;
23
:
5301
15
.
29
Li
QM
,
Tep
C
,
Yune
TY
, et al
. 
Opposite regulation of oligodendrocyte apoptosis by JNK3 and Pin1 after spinal cord injury
.
J Neurosci
2007
;
27
:
8395
404
.
30
Ding
Q
,
Huo
L
,
Yang
JY
, et al
. 
Down-regulation of myeloid cell leukemia-1 through inhibiting Erk/Pin 1 pathway by sorafenib facilitates chemosensitization in breast cancer
.
Cancer Res
2008
;
68
:
6109
17
.
31
Yu
C
,
Bruzek
LM
,
Meng
XW
, et al
. 
The role of Mcl-1 downregulation in the proapoptotic activity of the multikinase inhibitor BAY 43-9006
.
Oncogene
2005
;
24
:
6861
9
.
32
Kerbel
RS
. 
Tumor angiogenesis
.
N Engl J Med
2008
;
358
:
2039
49
.
33
Wu
G
,
Fang
YZ
,
Yang
S
,
Lupton
JR
,
Turner
ND
. 
Glutathione metabolism and its implications for health
.
J Nutr
2004
;
134
:
489
92
.
34
Mehdi
MZ
,
Azar
ZM
,
Srivastava
AK
. 
Role of receptor and nonreceptor protein tyrosine kinases in H2O2-induced PKB and ERK1/2 signaling
.
Cell Biochem Biophys
2007
;
47
:
1
10
.
35
Adams
JM
,
Cory
S
. 
Bcl-2-regulated apoptosis: mechanism and therapeutic potential
.
Curr Opin Immunol
2007
;
19
:
488
96
.
36
van Delft
MF
,
Wei
AH
,
Mason
KD
, et al
. 
The BH3 mimetic ABT-737 targets selective Bcl-2 proteins and efficiently induces apoptosis via Bak/Bax if Mcl-1 is neutralized
.
Cancer Cell
2006
;
10
:
389
99
.
37
Chen
L
,
Willis
SN
,
Wei
A
, et al
. 
Differential targeting of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apoptotic function
.
Mol Cell
2005
;
17
:
393
403
.
38
Chen
S
,
Dai
Y
,
Harada
H
,
Dent
P
,
Grant
S
. 
Mcl-1 down-regulation potentiates ABT-737 lethality by cooperatively inducing Bak activation and Bax translocation
.
Cancer Res
2007
;
67
:
782
91
.
39
Nguyen
M
,
Marcellus
RC
,
Roulston
A
, et al
. 
Small molecule obatoclax (GX15-070) antagonizes MCL-1 and overcomes MCL-1-mediated resistance to apoptosis
.
Proc Natl Acad Sci U S A
2007
;
104
:
19512
7
.
40
Mikula
M
,
Schreiber
M
,
Husak
Z
, et al
. 
Embryonic lethality and fetal liver apoptosis in mice lacking the c-raf-1 gene
.
EMBO J
2001
;
20
:
1952
62
.
41
Gollob
JA
,
Wilhelm
S
,
Carter
C
,
Kelley
SL
. 
Role of Raf kinase in cancer: therapeutic potential of targeting the Raf/MEK/ERK signal transduction pathway
.
Semin Oncol
2006
;
33
:
392
406
.
42
Brugarolas
J
. 
Renal-cell carcinoma-molecular pathways and therapies
.
N Engl J Med
2007
;
356
:
185
7
.
43
Meng
XW
,
Lee
SH
,
Dai
H
, et al
. 
Mcl-1 as a buffer for proapoptotic Bcl-2 family members during TRAIL-induced apoptosis: a mechanistic basis for sorafenib (Bay 43-9006)-induced TRAIL sensitization
.
J Biol Chem
2007
;
282
:
29831
46
.

Competing Interests

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.

Supplementary data