Proteins of the BCL2 family are key regulators of apoptosis. Their expression levels are frequently altered in cancers, enabling tumor cells to survive. To gain insight into the pathogenesis of hepatocellular carcinoma (HCC), we performed a comprehensive survey of the expression of the members of the BCL2 family in samples obtained from surgically resected HCCs. Here, we report the occurrence of a new molecular anomaly, consisting of a strong reduction in the expression of the proapoptotic protein BAD in HCC compared with surrounding nontumoral tissue. We investigate the function of BAD in a panel of HCC cell lines. Using gene overexpression and RNA interference, we show that BAD is involved in the cytotoxic effects of sorafenib, a multikinase blocker, which is currently the sole therapeutic drug effective for the treatment of HCC. Finally, we report that ABT-737, a compound that interacts with proteins of the BCL2 family and exhibits a BAD-like reactivity, sensitizes HCC cells toward sorafenib-induced apoptosis. Collectively, our findings indicate that BAD is a key regulator of apoptosis in HCC and an important determinant of HCC cell response to sorafenib. Mol Cancer Res; 8(8); 1116–25. ©2010 AACR.

Hepatocellular carcinoma (HCC) is a malignant tumor with a poor prognosis (1). Its progression is marked by the sequential accumulation of genetic/epigenetic abnormalities (2) that give rise to a set of common characteristics, among which is the ability to escape programmed cell death by apoptosis (3). The loss of sensitivity to apoptosis contributes to tumor progression and metastasis, and also determines how cancer cells respond to treatments (3, 4). Therefore, deciphering how apoptosis is regulated in HCC cells is an important goal to understand how these tumors develop and to improve their clinical handling.

Members of the BCL2 protein family function as positive or negative regulators of apoptosis, and they share a number of homology domains (Bcl2-homology domains BH1-BH4; ref. 5). Proapoptotic multidomain members, such as BAX and BAK, promote the release of apoptotic factors from mitochondria into the cytosol. They are regulated through interactions with antiapoptotic members of the BCL2 family, such as BCL2, BCLXL, or MCL1. A proapoptotic subset of BCL2 proteins characterized by a homology restricted to the BH3 domain of BCL2, and thus called the “BH3-only” members of the BCL2 family, plays a sentinel role in apoptosis regulation (5, 6). BH3-only members of the BCL2 family can either directly activate BAX and BAK or sensitize cells to apoptosis by neutralizing the antiapoptotic members of the BCL2 family (5, 6). The balance that results from the interactions between proapoptotic and antiapoptotic members of the BCL2 family determines the sensitivity of the cell to apoptosis (5, 6).

Changes in the expression levels and activation of one or more members of the BCL2 family occur in most types of tumors (7-10). Two antiapoptotic members of the BCL2 family, BCLXL and MCL1, have been shown to be expressed at increased levels in HCC (4, 11). Although the reported changes probably significantly contribute to hepatocarcinogenesis, no study has yet examined the complete BCL2 protein family. It remains unclear to what extent the expression of the other members of the BCL2 family of proteins changes in HCC. In particular, little is known about the variations in the expression of the proapoptotic members of the BH3-only family of proteins, although they have recently emerged as an important barrier against malignant transformation (10). A recent report indicates that BID, a member of the BH3-only subfamily, is downregulated in a subset of HCC appearing in the context of viral hepatitis (12). Whether the expression of other members of the BCL2 family of proteins is similarly altered in HCC remains unclear.

To address this issue, in the present study, we performed a survey of the expression of proteins from the BCL2 family, starting from surgically resected samples. We report the frequent occurrence of a decrease in the expression levels of BAD, a proapoptotic BH3-only member of the BCL2 family, in HCC. We examined the relevance of this decrease and analyzed its therapeutic implications.

Samples

Fourteen pairs of samples consisting of HCC/matched nontumor tissue were obtained from surgical resections done at Amiens University Hospital, France, between August 2005 and February 2009. All patients presenting with resectable HCC were included in this study. A detailed pathologic assessment of the tumor as well as nontumor tissues was done. A summary of the histologic observations and the baseline characteristics of the patients are provided in Supplementary Table S1. Sixteen control liver tissue samples were also obtained: the corresponding samples, hereby called “normal,” were obtained from surgical resections done following a diagnosis of hepatic metastasis of colorectal cancer. All samples were kept frozen at −80°C. The corresponding protocol was approved by the Comité de Protection des Personnes 2 Nord-Ouest.

Cell culture and reagents

All cell lines were cultured and characterized as indicated in Supplementary Material. Sorafenib (BAY49-4006) was kindly provided by Bayer Healthcare. ABT-737 and its inactive enantiomer were kindly provided by Abbott. All inhibitors were kept as stocks in DMSO at −20°C.

Western blots

Liver samples (minimal volume of 1 cm3 to prevent sampling artifacts) were homogenized in Laemmli buffer. Complete cell extracts were prepared in radioimmunoprecipitation assay buffer. Proteins were precipitated, loaded on SDS-PAGE, and transferred to nitrocellulose membranes using standard procedures. Antibodies used are described in Supplementary Material. Enhanced chemiluminescence reaction was used for revelation. Western blots were scanned and quantified using the software ImageJ (NIH).

Quantitative PCR

Total RNA was extracted using RNeasy Mini Kit (Qiagen) and reverse-transcribed using High Capacity cDNA Reverse Transcription kit and random hexamer (Applied Biosystems). Amplification was done with the TaqMan Universal PCR master Mix on an ABI 7900HT Sequence Detection System (Applied Biosystems) using primers and probe sets for BAD and human β-actin/glyceraldehyde 3-phosphate dehydrogenase (TaqMan Gene Expression Assay, Applied Biosystems).

RNA interference

Silencer select validated siRNA directed against BAD (si1861 and si1862) and Silencer negative control (am4635 and am4637siRNAs) were purchased from Applied Biosystems. Transfections were done using the siPORT-neoFX reagent (Applied Biosystems) and Opti-MEM transfection medium (Invitrogen), according to the manufacturer's instructions.

Fluorescence microscopy

Cells grown on glass coverslips were fixed with 3.7% paraformaldehyde, permeabilized with 0.01% Triton X-100, and stained with relevant antibodies. Nuclei were stained with 4′,6-diamidino-2-phenylindole. Coverslips were mounted in Mowiol (Calbiochem) and observed with a Nikon Eclipse TE2000U microscope equipped with a plan APO VC 60X/1.40 objective under oil immersion. Images were processed and superposed with the Photoshop software (Adobe).

Viability assays

Trypan blue exclusion assays were performed by the use of an automated cell counter Countess (Invitrogen), using the manufacturer's protocol. For clonogenicity assays, Huh7 cells (100 cells per 15-mm diameter dish) were incubated for 10 days under the indicated conditions. Cells were stained with Giemsa, and the number of clones with more than 20 cells is presented.

Statistical analyses

Wilcoxon unpaired test, Pearson r coefficient, and Student's t test were used as indicated. A value of P < 0.05 was considered as threshold for significance.

A subset of HCCs exhibit a dramatic reduction in BAD expression

To better understand how the survival of HCC cells is regulated, we analyzed the expression of the proteins of the BCL2 family in 14 tumor samples and matched nontransformed tissue surrounding the tumors (hereby called “nontumoral tissue”; Fig. 1A). We noticed large variations in the expression levels of some of the members of the BCL2 family. Setting a 3-fold change in expression as threshold, we noted an alteration in the expression levels of the proteins BAD, BID, BNIP3, BAX and BCLXL in more than one tumor (Table 1). The most frequent alteration that we observed was a strong reduction in the expression of BAD, a proapoptotic BH3-only member of the BCL2 family (Table 1): in 50.0% of the tumors examined (n = 7), BAD protein levels sharply decreased compared with nontumoral tissue. A reduction in the expression of two other proapoptotic proteins, BID and BAX, was also noticed, although at a lower frequency (Table 1). In contrast, BNIP3, another BH3-only protein of the BCL2 family, was frequently overexpressed (Table 1). None of the other proteins of the BCL2 family showed altered levels of expression: the proteins were expressed at stable levels (e.g., BAK or BIM), were found overexpressed in a single tumor (BCL2), or could not be detected at the protein level in samples obtained from normal liver or HCC.

FIGURE 1.

Variations in the expression levels of proteins of the BCL2 family in HCC compared with nontumoral tissue. A, three representative tumors (T) presenting with a decrease in their expression of BAD compared with the corresponding nontumoral (NT) tissues. The indicated markers were analyzed by immunoblotting. B, semiquantitative analysis of the expression of the main proteins of the BCL2 family in HCC. Quantitation was done as indicated in the text, and matched NT/T samples were compared with surgical samples from normal liver (N). Results are expressed in arbitrary units (A.U.) and presented as box plots. *, P < 0.005 compared with normal tissue; #, P < 0.005 compared with nontumoral tissue.

FIGURE 1.

Variations in the expression levels of proteins of the BCL2 family in HCC compared with nontumoral tissue. A, three representative tumors (T) presenting with a decrease in their expression of BAD compared with the corresponding nontumoral (NT) tissues. The indicated markers were analyzed by immunoblotting. B, semiquantitative analysis of the expression of the main proteins of the BCL2 family in HCC. Quantitation was done as indicated in the text, and matched NT/T samples were compared with surgical samples from normal liver (N). Results are expressed in arbitrary units (A.U.) and presented as box plots. *, P < 0.005 compared with normal tissue; #, P < 0.005 compared with nontumoral tissue.

Close modal
Table 1.

Summary of the variations in the expression of proteins of the BCL2 family

NameVariation% Tumors (n)
BAD Decrease 50.0% (7) 
BNIP3 Increase 50.0% (7) 
BID Decrease 35.7% (5) 
BCLXL Increase 28.6% (4) 
BAX Decrease 21.4% (3) 
NameVariation% Tumors (n)
BAD Decrease 50.0% (7) 
BNIP3 Increase 50.0% (7) 
BID Decrease 35.7% (5) 
BCLXL Increase 28.6% (4) 
BAX Decrease 21.4% (3) 

NOTE: Variations are ranked according to their frequency in the present study. We only report those that were found in more than one tumor.

To evaluate the changes in the expression of the main members of the BCL2 family of proteins in a semiquantitative fashion, a densitometric analysis of the signal measured for each protein was done. The corresponding results are presented in Fig. 1B (results are given in arbitrary units). Median intensity for BAD was 46.9 (95% confidence interval, 34.3–59.0) in normal livers versus 2.5 (95% confidence interval, 1–13.4) in tumors (P < 0.005, Wilcoxon test). Nontumoral tissues also exhibited reduced expression levels of BAD: 16.1 (95% confidence interval, 6.9–48.4) compared with normal liver, although this reduction did not reach statistical significance compared with normal liver samples. This approach confirmed the occurrence of important variations in BAD expression in HCC, a finding that had, to our knowledge, hitherto not been reported.

BAD is an unstable protein in HCC cells

To examine how the expression of BAD is regulated in HCC, we compared BAD mRNA levels in tumors and nontumoral tissues from three patients exhibiting strongly reduced expression levels of BAD: we found no reduction in the levels of BAD mRNA in these tumors by quantitative PCR (Fig. 2A), suggesting that posttranscriptional regulation mainly accounts for the disappearance of the BAD protein in HCC. Based on this observation, we decided to examine the stability of BAD in a panel of HCC cells. We treated four human hepatoma cell lines, PRL/PRF5, Hep3B, Huh7, and HepG2, either with chloroquine (100 μmol/L), a weak base that prevents lysosomal proteolysis, or the proteasome inhibitor MG132 (5 μmol/L). In all HCC cell lines, a 6-hour treatment with MG132 resulted in a near 3-fold increase in the expression of BAD, indicating that the basal half-life of BAD in HCC cells is around 1 hour (Fig. 2B; Supplementary Fig. S1). In contrast, protein levels of BCLXL, taken as control, remained stable (Supplementary Fig. S1). Therefore, in HCC cells, BAD is a short-lived protein that encounters continuous proteasomal degradation.

FIGURE 2.

Posttranscriptional regulation of BAD in HCC. A, quantitative analysis of BAD mRNA levels in HCC. BAD mRNA levels were analyzed in three tumors with decreased protein levels of BAD. Results are normalized to β-actin mRNA and are expressed as a tumor/nontumoral tissue ratio (results are average of two experiments). B, BAD protein exhibits a rapid turnover through proteasomal degradation. Various hepatoma cells were maintained for 6 h in the presence of chloroquine (Chlor; 100 μmol/L) or MG132 (5 μmol/L) and analyzed by immunoblotting. BAD expression is presented as mean value of three independent experiments. *, P < 0.05 compared with control.

FIGURE 2.

Posttranscriptional regulation of BAD in HCC. A, quantitative analysis of BAD mRNA levels in HCC. BAD mRNA levels were analyzed in three tumors with decreased protein levels of BAD. Results are normalized to β-actin mRNA and are expressed as a tumor/nontumoral tissue ratio (results are average of two experiments). B, BAD protein exhibits a rapid turnover through proteasomal degradation. Various hepatoma cells were maintained for 6 h in the presence of chloroquine (Chlor; 100 μmol/L) or MG132 (5 μmol/L) and analyzed by immunoblotting. BAD expression is presented as mean value of three independent experiments. *, P < 0.05 compared with control.

Close modal

Sorafenib, a multikinase inhibitor, activates BAD in HCC cells

Among the BH3-only family of protein, BAD is characterized by its proapoptotic effect achieved through the neutralization of antiapoptotic proteins such as BCLXL (13). This aspect of BAD function is under a stringent negative regulation by phosphorylation: Upon trophic factor signaling, prosurvival kinase pathways such as the RAF-MEK-ERK cascade and PKA and PKB kinases negatively regulate the apoptotic potency of BAD (13-19). On this basis, we decided to examine how BAD expression might regulate the survival of HCC cells. Sorafenib, a multikinase inhibitor, is at the moment the sole pharmaceutical therapy that is effective against HCC (20, 21), and it exerts potent proapoptotic effects on HCC cells (22). We anticipated that a part of the cytotoxic effects of sorafenib might come from its activation of BAD. To examine this possibility, we analyzed BAD expression levels and phosphorylation on Ser75, an important residue targeted by the RAF-MEK-ERK kinase pathway, a target of sorafenib (17-19), in HCC cells. We found that sorafenib increases BAD expression and prevents its inhibitory phosphorylation on Ser75 in Huh7 cells (Fig. 3A). The regulatory effect of sorafenib on BAD expression levels was at least partially accounted for by transcriptional mechanisms because increased levels of BAD mRNA were detected under these conditions (Fig. 3B). Because the RAF kinases are a target of sorafenib, we decided to examine the contribution of MEK inhibition to the effect of sorafenib on BAD. The chemical inhibitor U0126 and the bacterial toxin lethal factor purified from Bacillus anthracis were applied on HCC cells at doses blocking ERK phosphorylation (Supplementary Fig. S2). However, none of these strategies resulted in an increase in BAD expression in HCC cells (Supplementary Fig. S2), suggesting that the inhibition of MEK alone cannot account for the effects of sorafenib on BAD in HCC. We concluded that sorafenib activates BAD in HCC cells by regulating its expression levels and preventing its inhibition by phosphorylation.

FIGURE 3.

Sorafenib activates BAD in HCC. A, sorafenib modulates the expression of BAD and its activation in HCC cells. Huh7 cells were exposed to the indicated concentrations of sorafenib for 18 h, and were analyzed for their expression of BAD and its phosphorylated Ser75 form by immunoblotting. Results shown are representative of three independent experiments. B, quantitative analysis of BAD mRNA levels in HCC cells treated with sorafenib. Huh7 cells were treated for 18 h with sorafenib, and mRNA levels of BAD were analyzed by quantitative PCR. Results are normalized to glyceraldehyde 3-phosphate dehydrogenase mRNA (results are average from two experiments).

FIGURE 3.

Sorafenib activates BAD in HCC. A, sorafenib modulates the expression of BAD and its activation in HCC cells. Huh7 cells were exposed to the indicated concentrations of sorafenib for 18 h, and were analyzed for their expression of BAD and its phosphorylated Ser75 form by immunoblotting. Results shown are representative of three independent experiments. B, quantitative analysis of BAD mRNA levels in HCC cells treated with sorafenib. Huh7 cells were treated for 18 h with sorafenib, and mRNA levels of BAD were analyzed by quantitative PCR. Results are normalized to glyceraldehyde 3-phosphate dehydrogenase mRNA (results are average from two experiments).

Close modal

BAD determines the sensitivity of HCC cells to the cytotoxic effects of sorafenib

To examine the role of BAD in the toxicity of sorafenib in HCC, we decided to either overexpress or reduce the expression of this protein. Sorafenib-induced cytotoxicity in HCC cells was estimated by measuring chromatin condensation and trypan blue uptake, and by the use of a clonogenicity assay. Using these assays, we consistently observed a reduction in the viability of Huh7 cells exposed to sorafenib at concentrations that block intracellular phosphorylations (Supplementary Fig. S3). Stable cell lines derived from Huh7 cells overexpressing BAD were found to exhibit a striking 5-fold increase in their sensitivity to sorafenib-induced cytotoxicity measured by determining the mean of the percentage of cells with condensed chromatin (P < 0.05, Student's t test; Fig. 4A). Simultaneously, their clone-forming ability was strikingly reduced upon sorafenib exposure, even at the lowest doses examined (Fig. 4B). Conversely, an 80% reduction in the expression of BAD that was achieved using a siRNA knockdown strategy produced a 40% decrease in the number of cells with condensed chromatin after sorafenib treatment (P < 0.05, Student's t test; Fig. 4C). These results suggest that BAD plays an important role in sorafenib cytotoxicity in the Huh7 human HCC cell line. To extend this conclusion to various other HCC cell lines, we tested the existence of a possible correlation between the expression levels of BAD and the cytotoxicity of sorafenib on our panel of HCC cells in cultures (Fig. 4D). Interestingly, a strong correlation was found between the levels of expression of BAD in the four cell lines examined and sorafenib-induced cytotoxicity measured by the percentage of trypan blue uptake (Pearson r = 0.954). The cell lines with the highest levels of expression of BAD (PRL/PRF5 and Hep3B) were found approximately two times more sensitive to sorafenib than those with lower expression levels (Huh7 and HepG2) in terms of cytotoxicity (Fig. 4D). No such correlation could be established with the other members of the BCL2 family such as MCL1, BCLXL, or BIM (data not shown). Collectively, these results suggest that BAD is an important effector of sorafenib-induced cytotoxicity in HCC cells.

FIGURE 4.

BAD regulates the sensitivity of HCC cells to the cytotoxic effects of sorafenib. A, overexpression of BAD sensitizes HCC cells to sorafenib-induced cytotoxicity. Subclones derived from Huh7 cells and stably overexpressing green fluorescent protein (GFP) or GFP-BAD were established and characterized by immunoblotting. The corresponding clones were exposed to sorafenib (10 μmol/L, 18 h). Cells with condensed chromatin were identified after 4′,6-diamidino-2-phenylindole staining, and the percentage of apoptotic cells is shown (results were obtained in triplicate, with 150 cells counted each time). *, P < 0.05 compared with GFP cells exposed to sorafenib. B, overexpression of BAD reduces the clone-forming ability of Huh7 cells. Cell viability of Huh7 overexpressing GFP or GFP-BAD was estimated in a clonogenicity assay where different concentrations of sorafenib were tested. Results shown are the mean of three independent experiments. C, BAD siRNA protects Huh7 cells from sorafenib-induced cytotoxicity. Huh-7 cells were treated for 48 h with control or a mixture of two siRNAs targeting BAD, and their BAD protein content was analyzed by immunoblotting. Results from a representative experiment done in triplicate are presented, with each condition based on at least 100 cells counted on random fields; *, P < 0.05 compared with control siRNA C1 and C2. D, the expression levels of BAD correlate with sorafenib cytotoxicity in a panel of HCC cell lines. A correlation was established between the expression levels of BAD (arbitrary units) and the percentage of cells presenting a positive uptake of trypan blue after 18 h treatment with 10 μmol/L sorafenib (Pearson coefficient r = 0.954).

FIGURE 4.

BAD regulates the sensitivity of HCC cells to the cytotoxic effects of sorafenib. A, overexpression of BAD sensitizes HCC cells to sorafenib-induced cytotoxicity. Subclones derived from Huh7 cells and stably overexpressing green fluorescent protein (GFP) or GFP-BAD were established and characterized by immunoblotting. The corresponding clones were exposed to sorafenib (10 μmol/L, 18 h). Cells with condensed chromatin were identified after 4′,6-diamidino-2-phenylindole staining, and the percentage of apoptotic cells is shown (results were obtained in triplicate, with 150 cells counted each time). *, P < 0.05 compared with GFP cells exposed to sorafenib. B, overexpression of BAD reduces the clone-forming ability of Huh7 cells. Cell viability of Huh7 overexpressing GFP or GFP-BAD was estimated in a clonogenicity assay where different concentrations of sorafenib were tested. Results shown are the mean of three independent experiments. C, BAD siRNA protects Huh7 cells from sorafenib-induced cytotoxicity. Huh-7 cells were treated for 48 h with control or a mixture of two siRNAs targeting BAD, and their BAD protein content was analyzed by immunoblotting. Results from a representative experiment done in triplicate are presented, with each condition based on at least 100 cells counted on random fields; *, P < 0.05 compared with control siRNA C1 and C2. D, the expression levels of BAD correlate with sorafenib cytotoxicity in a panel of HCC cell lines. A correlation was established between the expression levels of BAD (arbitrary units) and the percentage of cells presenting a positive uptake of trypan blue after 18 h treatment with 10 μmol/L sorafenib (Pearson coefficient r = 0.954).

Close modal

The compound ABT-737 potentiates the cytotoxic effects of sorafenib in HCC cells

Because the proteins of the BCL2 family play a crucial role in the response of cancer cells to a variety of chemotherapeutic agents, pharmaceutical strategies have attempted to directly interfere with the function of these proteins (23). ABT-737 is a recently developed compound that binds to and inhibits the antiapoptotic proteins of the BCL2 family with a specificity mimicking BAD (23, 24). Based on our previous observations indicating that BAD is a key player in the regulation of HCC cell survival, we reasoned that ABT-737 might be a potent adjunct to sorafenib to promote its cytotoxicity in HCC cells. We tested the cytotoxic effects of ABT-737 applied at various concentrations on HCC cells (Supplementary Fig. S4). To investigate the possible synergy between ABT-737 and sorafenib, we applied ABT-737 or its inactive enantiomer at subtoxic concentrations (5 μmol/L). When Huh7 cells were simultaneously exposed to ABT-737 and sorafenib, we observed a strong synergy between these two compounds in terms of apoptosis induction, estimated by condensed chromatin formation under microscopic examination (Fig. 5A), a clonogenicity assay (Fig. 5B), or with caspase-3 cleavage followed by immunoblotting (Fig. 5C). Similar results were obtained with HepG2 cells (Supplementary Fig. S5A and B), whereas various cell lines of nonliver origin were much less sensitive to sorafenib and ABT-737 (Supplementary Fig. S5C). The combined cytotoxic effect of ABT-737 and sorafenib on HCC cells clearly resulted from the mitochondrial action of these drugs: cytochrome c was released from the mitochondria of Huh7 cells as early as 3 hours after the exposure to ABT-737/sorafenib, but not after a single application of ABT-737 or sorafenib alone, as shown by immunoblotting (Fig. 5D) or immunofluorescence (Fig. 6). We concluded that ABT-737 potentiates the induction of apoptosis by sorafenib in HCC cells.

FIGURE 5.

ABT-737 potentiates sorafenib-induced cytotoxicity on HCC cells. A, quantification of the percentage of Huh7 cells with condensed chromatin. Sorafenib (10 μmol/L) was applied for 18 h, alone or in the presence of ABT-737 or its inactive enantiomer (Ctrl), both at 5 μmol/L. Results shown are from a representative experiment done in triplicate, with each condition based on at least 100 cells counted on random fields. *, P < 0.05 compared with all other conditions. B, clonogenicity. Huh7 cells were exposed to ABT-737 or its inactive enantiomer (Ctrl) at a concentration of 5 μmol/L, together with sorafenib at a subtoxic concentration (2 μmol/L). Results are from a single experiment done in triplicate. *, P < 0.05 compared with nontreated cells as well as cells treated with ABT-737 alone. C, caspase-3 cleavage. Cell lysates were prepared from Huh7 cells treated as described previously, and an immunoblot was done to label caspase-3. The arrows indicate full-length caspase-3, and its active, 19 kDa cleavage product. D, cytochrome c cytosolic content. Cytosolic and total extracts were prepared from Huh7 cells exposed to ABT-737 and/or sorafenib, and were analyzed for their cytochrome c content by immunoblotting.

FIGURE 5.

ABT-737 potentiates sorafenib-induced cytotoxicity on HCC cells. A, quantification of the percentage of Huh7 cells with condensed chromatin. Sorafenib (10 μmol/L) was applied for 18 h, alone or in the presence of ABT-737 or its inactive enantiomer (Ctrl), both at 5 μmol/L. Results shown are from a representative experiment done in triplicate, with each condition based on at least 100 cells counted on random fields. *, P < 0.05 compared with all other conditions. B, clonogenicity. Huh7 cells were exposed to ABT-737 or its inactive enantiomer (Ctrl) at a concentration of 5 μmol/L, together with sorafenib at a subtoxic concentration (2 μmol/L). Results are from a single experiment done in triplicate. *, P < 0.05 compared with nontreated cells as well as cells treated with ABT-737 alone. C, caspase-3 cleavage. Cell lysates were prepared from Huh7 cells treated as described previously, and an immunoblot was done to label caspase-3. The arrows indicate full-length caspase-3, and its active, 19 kDa cleavage product. D, cytochrome c cytosolic content. Cytosolic and total extracts were prepared from Huh7 cells exposed to ABT-737 and/or sorafenib, and were analyzed for their cytochrome c content by immunoblotting.

Close modal
FIGURE 6.

ABT-737 and sorafenib synergistically promote cytochrome c release from mitochondria of HCC cells. Huh7 cells exposed for 3 h to sorafenib (10 μmol/L) and/or ABT-737 (5 μmol/L) were labeled with TOM20 (green) to visualize their mitochondria, and cytochrome c was labeled in parallel (red). An overlay is presented in the right column. In the lower panel, arrowheads indicate cells with cytochrome c release. Bar, 25 μm.

FIGURE 6.

ABT-737 and sorafenib synergistically promote cytochrome c release from mitochondria of HCC cells. Huh7 cells exposed for 3 h to sorafenib (10 μmol/L) and/or ABT-737 (5 μmol/L) were labeled with TOM20 (green) to visualize their mitochondria, and cytochrome c was labeled in parallel (red). An overlay is presented in the right column. In the lower panel, arrowheads indicate cells with cytochrome c release. Bar, 25 μm.

Close modal

Molecular basis for the synergistic effect between ABT-737 and sorafenib on HCC cells

To explore the mechanism that accounts for the synergistic effect of ABT-737 and sorafenib on HCC cells, we analyzed the expression of the various members of the BCL2 family of proteins in the panel of HCC cell lines (Fig. 7). Cellular extracts were prepared from Huh7, HepG2, Hep3B, and PRL/PRF5 cell lines exposed for 3 hours to sorafenib and/or ABT-737. We noted that sorafenib altered the expression levels of some proapoptotic as well as antiapoptotic members of the BCL2 family. However, a strong increase in the expression levels of BAD was the most striking effect detected in the four cell lines treated with ABT-737 (Fig. 7). A reduction in the expression of the prosurvival protein MCL1 combined with an increase in the expression of the proapoptotic BIM were also found in the Huh7 cell line, in accordance with the previous report by Liu et al. (22), pointing to MCL1 as a target for sorafenib. However, the expression levels of MCL1 and BIM were stable over this short period in the other three cell lines (Fig. 7). We concluded that the sensitizing action of ABT-737 toward sorafenib correlates with the ability of this compound to increase the expression and apoptotic potency of BAD. Finally, we examined the effect of sorafenib and ABT-737 on the activation of the proapoptotic protein BAX, a core component of the mitochondrial membrane permeabilization machinery (5). An increase in BAX immunoreactivity toward the monoclonal antibody 6A7, raised against the amino-terminus of BAX, was found in Huh7 cells treated simultaneously with sorafenib and ABT-737 for 3 hours, showing that BAX was readily activated under these conditions (Supplementary Fig. S6). We concluded that the experimental modulation of BCL2 expression and activation afforded by ABT-737 constitutes a promising strategy for the potentiation of apoptosis induction of HCC cells and that BAD is a possible therapeutic target for this type of cancer.

FIGURE 7.

ABT-737 and sorafenib regulate the expression and activity of proteins of the BCL2 family in HCC cells. Cellular extracts were prepared from Huh7, HepG2, Hep3B, or PRL/PRF5 cells exposed to sorafenib (10 μmol/L) and/or ABT-737 (10 μmol/L) for 3 h. Expression levels of the indicated proteins were analyzed by immunoblotting.

FIGURE 7.

ABT-737 and sorafenib regulate the expression and activity of proteins of the BCL2 family in HCC cells. Cellular extracts were prepared from Huh7, HepG2, Hep3B, or PRL/PRF5 cells exposed to sorafenib (10 μmol/L) and/or ABT-737 (10 μmol/L) for 3 h. Expression levels of the indicated proteins were analyzed by immunoblotting.

Close modal

Reduced sensitivity to apoptosis is a pivotal event in tumor progression (3). A decrease in the susceptibility to apoptosis can theoretically be achieved either by overexpression of antiapoptotic members of the BCL2 family or downregulation of their proapoptotic counterparts. Here, we report a new mechanism capable of preventing apoptosis in HCC cells, involving a reduction in the expression of BAD, a proapoptotic protein of the BCL2 family. Our findings are consistent with the emerging view that the BH3-only proteins are key regulators of apoptosis, whose expression is often altered in tumor cells (10). Recently, BID, another member of the subfamily of BH3-only proteins, was shown to be downregulated in a subset of HCC occurring in the context of viral hepatitis (12). Based on our results, we suggest that the alteration of the expression of selected BH3-only proteins is an important event in a subset of HCC. Because our study is based on a small population of resected HCC, larger cohorts will be required to precisely estimate the frequency of each molecular anomaly and relate them to clinical parameters.

In this study, we explored the role of BAD in cell death, taking into account that BAD was reported to exert an important regulatory role in this process in normal liver cells (25). We observed that sorafenib, a kinase inhibitor that is currently the sole medical treatment with shown efficacy on HCC, partially exerts its proapoptotic effects on HCC cells through BAD. Sorafenib increases the expression of BAD and at the same time prevents its inhibitory phosphorylation, thereby sensitizing HCC cells to apoptosis. Although sorafenib is essentially characterized as an inhibitor of the RAF kinases, we found that the sole inhibition of the RAF-MEK-ERK pathway cannot account for the effect of sorafenib on BAD. Chemical and enzymatic inhibitors of the MEK kinases applied on HCC cells did not reproduce the effect of sorafenib on BAD and did not exert a significant cytotoxic effect on HCC cells (data not shown). Interestingly, several kinases and phosphatases regulate BAD in a tiered fashion, and BAD probably serves as a signaling node, integrating essential pathways to regulate the apoptotic program (13, 18). The identification of the kinase(s) that regulate BAD and are the target(s) of sorafenib in HCC is therefore a complex task that will require further investigations. In addition, more studies will be required to examine the functional consequences of the reduction in expression of BAD that we report in HCC. Beyond apoptosis regulation, BAD was recently reported to modulate the cell cycle, autophagy, or glucose metabolism in different cell types (13). Whereas our observations indicate that BAD is a key player in the regulation of apoptosis in HCC cells, further studies examining the function of BAD in liver carcinogenesis are clearly required.

How could our findings help improve the current handling of HCC? Although sorafenib is effective for the treatment of HCC, patients respond in different ways to this treatment, and there are no good parameters that can be used to predict which patients will benefit the most from this treatment (26, 27). Assuming that the death of tumor cells is important to achieve HCC regression, we propose that the exploration of BAD expression within the tumor cells might help identify these patients. A more remote application of our findings might be the improvement of therapeutic schemes based on sorafenib. Strategies based on the interference with proteins of the BCL2 family have recently emerged as promising adjuncts to oncogenic kinase inhibitors for the treatment of different cancers (28). Our observations on ABT-737 constitute, to our knowledge, the first report on the use of a BCL2 inhibitor in HCC. Our results suggest that interference with proteins of the BCL2 family might offer clinical benefits in these tumors. Future therapeutic strategies aiming to restore the function of BAD in the cancer cell might therefore help improve the treatment of HCC.

J-C. Barbare: consultant, Bayer France. Other authors have reported no conflict of interest.

We thank Momar Diouf, Jean-Paul Galmiche, and Zuzana Saidak for helpful comments; Bayer for the gift of sorafenib; Abbott for the gift of ABT-737; and the Ligue Nationale contre le Cancer-Comité de la Somme for financial support.

Grant Support: Ligue Nationale contre le Cancer, Comité de la Somme. The funder had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

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.

1
El-Serag
HB
,
Rudolph
KL
. 
Hepatocellular carcinoma: epidemiology and molecular carcinogenesis
.
Gastroenterology
2007
;
132
:
2557
76
.
2
Aravalli
RN
,
Steer
CJ
,
Cressman
ENK
. 
Molecular mechanisms of hepatocellular carcinoma
.
Hepatology
2008
;
48
:
2047
63
.
3
Hanahan
D
,
Weinberg
RA
. 
The hallmarks of cancer
.
Cell
2000
;
100
:
57
70
.
4
Fabregat
I
. 
Dysregulation of apoptosis in hepatocellular carcinoma cells
.
World J Gastroenterol
2009
;
15
:
513
20
.
5
Youle
RJ
,
Strasser
A
. 
The BCL-2 protein family: opposing activities that mediate cell death
.
Nat Rev Mol Cell Biol
2008
;
9
:
47
59
.
6
Leber
B
,
Lin
J
,
Andrews
DW
. 
Embedded together: the life and death consequences of interaction of the Bcl-2 family with membranes
.
Apoptosis
2007
;
12
:
897
911
.
7
Certo
M
,
Del Gaizo Moore
V
,
Nishino
M
, et al
. 
Mitochondria primed by death signals determine cellular addiction to antiapoptotic BCL-2 family members
.
Cancer Cell
2006
;
9
:
351
65
.
8
Letai
AG
. 
Diagnosing and exploiting cancer's addiction to blocks in apoptosis
.
Nat Rev Cancer
2008
;
8
:
121
32
.
9
Yip
KW
,
Reed
JC
. 
Bcl-2 family proteins and cancer
.
Oncogene
2008
;
27
:
6398
406
.
10
Frenzel
A
,
Grespi
F
,
Chmelewskij
W
,
Villunger
A
. 
Bcl2 family proteins in carcinogenesis and the treatment of cancer
.
Apoptosis
2009
;
14
:
584
96
.
11
Takehara
T
,
Liu
X
,
Fujimoto
J
,
Friedman
SL
,
Takahashi
H
. 
Expression and role of BclxL in human hepatocellular carcinomas
.
Hepatology
2001
;
34
:
55
61
.
12
Simonin
Y
,
Disson
O
,
Lerat
H
, et al
. 
Calpain activation by hepatitis C virus proteins inhibits the extrinsic apoptotic signaling pathway
.
Hepatology
2009
;
50
:
1370
9
.
13
Danial
NN
. 
BAD: undertaker by night, candyman by day
.
Oncogene
2009
;
27
:
S53
70
.
14
Zha
J
,
Harada
H
,
Yang
E
,
Jockel
J
,
Korsmeyer
SJ
. 
Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14-3-3 not BCL-X(L)
.
Cell
1996
;
87
:
619
28
.
15
Wang
HG
,
Rapp
UR
,
Reed
JC
. 
Bcl-2 targets the protein kinase Raf-1 to mitochondria
.
Cell
1996
;
87
:
629
38
.
16
Datta
SR
,
Ranger
AM
,
Lin
MZ
, et al
. 
Survival factor-mediated BAD phosphorylation raises the mitochondrial threshold for apoptosis
.
Dev Cell
2002
;
3
:
631
43
.
17
Eisenmann
KM
,
VanBrocklin
MW
,
Staffend
NA
,
Kitchen
SM
,
Koo
HM
. 
Mitogen-activated protein kinase pathway-dependent tumor-specific survival signaling in melanoma cells through inactivation of the proapoptotic protein bad
.
Cancer Res
2003
;
63
:
8330
7
.
18
She
QB
,
Solit
DB
,
Ye
Q
,
O'Reilly
KE
,
Lobo
J
,
Rosen
N
. 
The BAD protein integrates survival signaling by EGFR/MAPK and PI3K/Akt kinase pathways in PTEN-deficient tumor cells
.
Cancer Cell
2005
;
8
:
287
97
.
19
Fueller
J
,
Becker
M
,
Sienerth
AR
,
Fischer
A
,
Hotz
C
,
Galmiche
A
. 
C-RAF activation promotes BAD poly-ubiquitylation and turn-over by the proteasome
.
Biochem Biophys Res Commun
2008
;
370
:
552
6
.
20
Llovet
JM
,
Ricci
S
,
Mazzaferro
V
, et al
. 
Sorafenib in advanced hepatocellular carcinoma
.
N Engl J Med
2008
;
359
:
378
90
.
21
Zhang
J
,
Yang
PL
,
Gray
NS
. 
Targeting cancer with small molecule kinase inhibitors
.
Nat Rev Cancer
2009
;
9
:
28
39
.
22
Liu
L
,
Cao
Y
,
Chen
C
, et al
. 
Sorafenib blocks the RAF/MEK/ERK pathway, inhibits tumor angiogenesis, and induces tumor cell apoptosis in hepatocellular carcinoma model PLC/PRF/5
.
Cancer Res
2006
;
66
:
11851
8
.
23
Chonghaile
TN
,
Letai
A
. 
Mimicking the BH3 domain to kill cancer cells
.
Oncogene
2008
;
27
:
S149
57
.
24
Oltersdorf
T
,
Elmore
SW
,
Shoemaker
AR
, et al
. 
An inhibitor of Bcl-2 family proteins induces regression of solid tumours
.
Nature
2005
;
435
:
677
81
.
25
Danial
NN
,
Gramm
CF
,
Scorrano
L
, et al
. 
BAD and glucokinase reside in a mitochondrial complex that integrates glycolysis and apoptosis
.
Nature
2003
;
424
:
952
6
.
26
Abou-Alfa
GK
. 
Selection of patients with hepatocellular carcinoma for sorafenib
.
J Natl Compr Canc Netw
2009
;
7
:
397
403
.
27
Abou-Alfa
GK
,
Schwartz
L
,
Ricci
S
, et al
. 
Phase II study of sorafenib in patients with advanced hepatocellular carcinoma
.
J Clin Oncol
2006
;
24
:
4293
300
.
28
Cragg
MS
,
Harris
C
,
Strasser
A
,
Scott
CL
. 
Unleashing the power of inhibitors of oncogenic kinases through BH3 mimetics
.
Nat Rev Cancer
2009
;
9
:
321
6
.