ORIGINAL ARTICLE
See related commentary on pg 315
RAF Inhibition Overcomes Resistance to TRAIL-Induced Apoptosis in Melanoma Cells Anja Berger1, Sandra-Annika Quast1, Michael Plo¨tz1, Nicholas-Frederik Kuhn1, Uwe Trefzer1 and Ju¨rgen Eberle1 Mutated BRAF represents a critical oncogene in melanoma, and selective inhibitors have been approved for melanoma therapy. However, the molecular consequences of RAF inhibition in melanoma cells remained largely elusive. Here, we investigated the effects of the pan-RAF inhibitor L-779,450, which inhibited cell proliferation both in BRAF-mutated and wild-type melanoma cell lines. It furthermore enhanced apoptosis in combination with the death ligand tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) and overcame TRAIL resistance in melanoma cells. Enhanced apoptosis coincided with activation of mitochondrial pathways, seen by loss of mitochondrial membrane potential and release of cytochrome c, Smac (second mitochondria–derived activator of caspases), and apoptosis-inducing factor (AIF). Subsequently, caspase-9 and -3 were activated. Apoptosis induction by L-779,450/TRAIL was prevented by Bcl-2 overexpression and was dependent on Bax. Thus, activation of Bax by L-779,450 alone was demonstrated by Bax conformational changes, whereas Bak was not activated. Furthermore, the BH3-only protein Bim was upregulated in response to L-779,450. The significant roles of Smac, Bax, and Bim in this setting were proven by small interfering RNA (siRNA)-mediated knockdown experiments. L-779,450 also resulted in morphological changes indicating autophagy confirmed by the autophagy marker light chain 3-II (LC3-II). The pro-apoptotic effects of L-779,450 may explain the antitumor effects of RAF inhibition and may be considered when evaluating RAF inhibitors for melanoma therapy. Journal of Investigative Dermatology (2014) 134, 430–440; doi:10.1038/jid.2013.347; published online 19 September 2013
INTRODUCTION The high mortality of metastatic melanoma strongly relates to its pronounced resistance to chemotherapy as well as to an antitumor immune response (Garbe and Leiter, 2009). This is explained by defects in pro-apoptotic signaling, suggesting the overcoming of melanoma apoptosis deficiency as a predominant therapeutic target (Eberle et al., 2007). Mitogen-activated protein kinase (MAPK) pathways, initiated via Ras and RAF, have important roles in tumor development (Gray-Schopfer et al., 2005). The signaling cascade via MAPK/extracellular signal-regulated kinase (ERK) kinase (MEK) and ERK leads to phosphorylation of multiple target proteins, which drive the regulation of gene expression, cell differentiation, proliferation, and survival (Inamdar et al., 1
Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charite´, Berlin, Germany
Correspondence: Ju¨rgen Eberle, Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charite´, Charite´platz 1, 10117 Berlin, Germany. E-mail:
[email protected] Abbreviations: AIF, apoptosis-inducing factor; FCS, fetal calf serum; MAPK, mitogen-activated protein kinase; MEK, MAPK/ERK kinase; pERK, phosphorylated extracellular signal-regulated kinase kinase; Smac, second mitochondria–derived activator of caspases; TRAIL, tumor necrosis factor– related apoptosis-inducing ligand; XIAP, X-linked inhibitor of apoptosis; WT, wild type Received 17 October 2012; revised 14 May 2013; accepted 26 May 2013; accepted article preview online 16 August 2013; published online 19 September 2013
430
Journal of Investigative Dermatology (2014), Volume 134
2010). Three RAF isoforms (A, B, and C) have been reported, which exert distinct roles in different cells (Maurer et al., 2011). For melanoma, activating mutations in N-Ras (10–25%) and BRAF (40–60%) appear to be of particular interest (Davies et al., 2002; Sullivan and Flaherty, 2012). The high frequency of the BRAF(V600E) mutation resulted in the development of selective BRAF inhibitors, partially already approved for melanoma therapy (Flaherty et al., 2010; Chapman et al., 2011; Hauschild et al., 2012; Long et al., 2012; Sosman et al., 2012). However, the significance of RAF inhibitors in the regulation of apoptosis in melanoma cells has not been sufficiently explored. In apoptosis control, extrinsic proapoptotic pathways are initiated by binding of death ligands as tumor necrosis factor-a, CD95L/FasL, and tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) to their respective death receptors, leading to the formation of death-inducing signaling complexes and activation of initiator caspase-8 (Krammer et al., 2007). On the other hand, intrinsic apoptosis pathways may be initiated by cellular dysregulation and DNA damage and may result in pro-apoptotic mitochondrial activation. Key events are depolarization of the mitochondrial membrane potential (Dcm) and mitochondrial outer membrane permeabilization, leading to the release of cytochrome c, Smac (second mitochondria–derived activator of caspases), and AIF (apoptosis-inducing factor). Whereas cytochrome c activates initiator caspase-9, Smac functions as an antagonist & 2014 The Society for Investigative Dermatology
A Berger et al. RAF in TRAIL Resistance
20
44 42
pERK
44 42
ERK
44 42 44 42
ERK
44 42
GAP DH
37
48 h
20 Apoptosis (%)
0
–
0.5
0.1
+
5
1
L-779
L-779
A-375 (V600E)
– +
10
SK-Mel-13
MeWo
10
10 5
0 L-779 (μM)
50
10
Mel-2a
Mel-HO-TS
0
pERK
L-779 (μM)
A-375
0
10 5
0 20
0 20
10 5
SK-Mel-28 (V600E)
Mel-HO (V600E)
*
10 5
*
40
0
20
10 5
40 20
SK-Mel-13 (V600E)
0
10 5
40 20
SK-Mel-103 (BRAF-WT)
*
*
0 72 h
Apoptosis (%)
20
A-375-TS (V600E)
Mel-HO-TS (V600E)
Mel-2a (V600E)
*
*
MeWo (BRAF-WT)
SK-Mel-147 (BRAF-WT)
Counts
ERK
Apoptosis (%)
44 42
LDH (x-fold)
MeWo (BRAF-WT)
A-375-TS (V600E)
A-375 (V600E)
kDa pERK
*
*
10
0 A-375
A-375TS
MelHO
Fluorescence intensity
2.5
A-375
2.5
2.5
A-375-TS
3.5
Mel-HO 0
2
2
0
3
2
0
Cell index
1.5
1.5 5
1
*
* 1
1
10 20
0.5
10
0.5
0
0.5
20
0 0
24
48
* 5
1.5 1
24
48
5
24
48
*
20
0.5
20 0
0 0
10
*
10
0.5
0 0
0 5 1
2
10 20
MeWO
0
2.5
5 1.5
1.5
SK-Mel-13
0
24
48
0
24
48
Time (h)
Figure 1. Antiproliferative effects by L-779,450. (a) Levels of phosphorylated extracellular signal-regulated kinase (pERK) and total ERK were determined by western blotting in human melanoma cell lines treated for 24 hours with L-779,450 (0.1–50 mM): A-375, A-375-TS (V600E), and MeWo (BRAF-wild type (WT)). Two independent experiments revealed comparable results; equal loading was proven by glyceraldehyde dehydrogenase (GAPDH). (b) Apoptosis (% of sub-G1 cells) was determined in response to L-779,450 (5–40 mM; 24 hours) in A-375, Mel-HO-TS, Mel-2a, MeWo, and SK-Mel-13. Cytotoxicity (lactate dehydrogenase (LDH) release) was determined in parallel (below). Here, values of non-treated cells were set to 1. (c) Apoptosis was also determined at 48 and 72 hours (L-779,450, 10 mM). (b, c) Means and SDs of triplicates; at least two independent experiments. (d) Cell cycle analyses of human melanoma cell lines: open graphs, L-779,450-treated (10 mM, 24 hours); filled graphs, dimethyl sulfoxide (DMSO)-treated controls. (e) Growth curves determined in real time of melanoma cell lines treated with L-779,450 (5–20 mM) were compared with controls (0). Means and SDs (at 48 hours) of triplicate determinations are shown; two independent experiments were performed. Statistical significance is indicated (*Po0.05), when comparing L-779,450 and DMSO-treated cells.
of cellular inhibitor of apoptosis proteins (e.g. X-linked inhibitor of apoptosis (XIAP) and survivin), which themselves prevent effector caspase activity (Tait and Green, 2010). Mitochondrial apoptosis pathways are critically controlled by the family of Bcl-2 proteins, which encloses anti-apoptotic (e.g. Bcl-2 and Mcl-1), pro-apoptotic multidomain (Bax and Bak), and pro-apoptotic BH3-only proteins (e.g. Bid and Bim) (Chipuk et al., 2010). Initiator caspases activated in both
pathways may drive the processing and activation of the main effector caspase-3, which itself cleaves various death substrates to set apoptosis into work (Fischer et al., 2003). Mitochondrial pathways may also be activated by death ligands via caspase-8mediated processing and activation of Bid (Li et al., 1998). The death ligand TRAIL represents an additional therapeutic strategy and selectively induces apoptosis in cancer cells via TRAIL receptors DR4 and DR5 (Newsom-Davis et al., 2009). www.jidonline.org
431
A Berger et al. RAF in TRAIL Resistance
In melanoma cells and tissue samples, we have previously shown functional activity of TRAIL receptors (Kurbanov et al., 2005); however, some melanoma cell lines revealed perma-
40
nent TRAIL resistance and others developed inducible resistance upon TRAIL treatment (Kurbanov et al., 2007). Inducible resistance was related to the downregulation of TRAIL
*
TRAIL –
sG1
+ Apoptosis (%)
30
Control TRAIL L779
sG1
*
*
*
*
A-375
*
*
0 10 Mel-HO (V600E)
0 20 Mel-HO-TS (V600E)
20
Mel-2a
*
10
LDH (fold change)
0
10 5
0 L-779 (μM)
10 0 A-375 (V600E)
0 10 A-375-TS (V600E)
Control
40 Mel-2a (V600E)
0
A-375-TS
2.5
10
5
+ TRAIL
*
20
10
10
0
0
TRAIL
40
0
24 h
48 h
1 L-779
0
24
A-375 (V600E)
20
*
– TRAIL
0.5 L-779/TRAIL
*
0
48 Time (h)
72 h
*
0
U0126 – +
3
Cell index
Apoptosis (%)
* MeWO (BRAFWT)
*
0
20 A-375
0 20 A-375-TS
0 20 Mel-HO
0
* – TRAIL
*
Apoptosis (%)
+ TRAIL
TRAIL 1 U0126/TRAIL
A-375 *
24 Time (h)
0
*
PLX4032 – +
A-375-TS *
20
Mel-HO
10 *
0 PLX4032 (μM)
432
U0126
Fluorescence intensity
20 MeWo
30
A-375
2
0 0 U0126 (μM)
0
3 A-375
10
0
3 A-375-TS
Journal of Investigative Dermatology (2014), Volume 134
10
0
10 Mel-HO
40
48
24
Control
+ TRAIL
10
L-779
TRAIL 1
40
0
Control
1.5
1.5
0 40
A-375-TS
2
Control
0.5
0
10
2.5
A-375
L-779/TRAIL 0 L-779 (μM)
0 10 SK-Mel-147 (BRAF-WT NRAS*)
30
2
+ TRAIL Cell index
Apoptosis (%)
– TRAIL
0 10 SK-Mel-103 (BRAF-WT NRAS*)
– TRAIL
*
DAPI-positive cells (%)
A-375
40
MeWo
15
0 40 MeWo (BRAF-WT)
A-375-TS
A-375
L779/TRAIL
0 L-779 (μM)
20
0 40 SK-Mel-13 (V600E)
0 20 SK-Mel-28 (V600E)
*
48
A Berger et al. RAF in TRAIL Resistance
receptors, initiator caspases, and pro-apoptotic Bcl-2 proteins (Zhang et al., 2006; Kurbanov et al., 2007), whereas the causes of permanent resistance in melanoma remained elusive. The correlation of MAPK activity and resistance to TRAILinduced apoptosis in melanoma cells had already been shown in a previous study when applying an MEK inhibitor (Zhang et al., 2003). Here, we prove the strong relation of RAF inhibition and apoptosis regulation in melanoma cells. As shown by small interfering RNA (siRNA)-mediated knockdown studies, the sensitization for TRAIL was particularly based on Bax activation and on upregulation of Bim in response to the inhibitor alone. This resulted in the activation of mitochondrial apoptosis pathways, particularly based on the release of Smac. RESULTS Direct effects of L-779,450
The pan-RAF inhibitor L-779,450 was applied for understanding the relations of RAF inhibition and apoptosis regulation in melanoma cells. Strong and dose-dependent phosphorylated ERK (pERK) downregulation was seen by L-779,450 in the BRAF(V600E)-mutated, TRAIL-sensitive melanoma cell line A-375, as well as in TRAIL-resistant A-375-TS. Reduced sensitivity of A-375-TS may be explained by its selection for TRAIL resistance. In contrast, only incomplete pERK downregulation was seen in the BRAF-wild-type (WT) melanoma cell line MeWo, even at high concentrations (Figure 1a). Downregulation of pERK largely remained without direct effect on apoptosis and cytotoxicity in melanoma cells up to 72 hours (Figure 1b and c). However, partial G1 arrest and depletion of S-phase cells were seen at 24 hours in four BRAF-mutated melanoma cell lines (A-375, Mel-HO, Mel-2a, and SK-Mel-28) as well as in their TRAIL-selected derivatives A-375-TS and Mel-HO-TS. G1 arrest was also seen in two N-Ras-mutated, BRAF-WT cell lines (SK-Mel-103 and SK-Mel-147), whereas another BRAFWT cell line (MeWo) and the BRAF-mutated cell line SK-Mel13 did not respond (Figure 1d). Treatment with L-779,450 resulted in dose-dependent decrease of cell numbers, as determined in real time (Figure 1e). Sensitization for death ligand–induced apoptosis
The sensitivity of melanoma cell lines for the death ligand TRAIL had been described in previous studies (Kurbanov et al., 2005, 2007; Berger et al., 2011). The effects of L-779,450 on TRAIL sensitivity were investigated here in melanoma cell lines with high TRAIL sensitivity (A-375 and SK-Mel-147),
moderate sensitivity (Mel-HO, SK-Mel-13, and SK-Mel-28), and permanent resistance (MeWo, Mel-2a, and SK-Mel-103), as well as in TRAIL-selected cell lines with acquired resistance (A-375-TS and Mel-HO-TS). Despite only moderate direct effects of L-779,450 on apoptosis, it strongly enhanced TRAILinduced apoptosis in sensitive melanoma cells and overruled TRAIL resistance in Mel-2a, SK-Mel-103, A-375-TS, and MelHO-TS. At 24 hours, 16–35% apoptosis induction was obtained, whereas cytotoxicity remained largely unaffected. BRAF-WT cells MeWo as well as BRAF-mutated cells SK-Mel-13 revealed no enhancement, not even at 72 hours after treatment (Figure 2a, b, and e). Morphologic changes characteristic for apoptosis became evident by bisbenzimide nuclear staining. Thus, nuclear fragmentation and chromatin condensation were seen at 24 hours in 18–30% of A-375 and A-375-TS cells (Figure 2c and d). The strong impact of induced apoptosis on cell proliferation was visualized by real-time cell analysis. Thus, complete detachment of cells was seen in A-375 and A-375-TS already at 36 hours in response to L-779,450/TRAIL (20 ng ml 1; Figure 2f). Treatment with the MEK inhibitor U0126 (20 mM) was applied for comparison and revealed a similar picture. Thus, it also enhanced TRAIL-induced apoptosis in A-375, A-375-TS, and Mel-HO but not in MeWo (Figure 2g–i). Also, treatment with the selective BRAF(V600E) inhibitor vemurafenib/PLX4032 resulted in comparable effects. Thus, PLX4032 similarly induced a G1 arrest and enhanced TRAIL-induced apoptosis in A-375, A-375-TS, and Mel-HO (Figure 2j and k). Activation of mitochondrial pro-apoptotic pathways
Involvement of caspases and Bid was investigated by monitoring activated cleavage products and loss of proforms, respectively. Processing of the extrinsic initiator caspase-8 and of Bid was already seen in A-375 and A-375-TS melanoma cells in response to TRAIL (16 hours), despite lacking TRAIL responsiveness of A-375-TS. This was evident by caspase-8 cleavage products (41 and 43 kDa) as well as by loss of caspase-8 and Bid proforms (57 and 22 kDa). This processing was, however, not enhanced by the combination with L-779,450 (Figure 3a). Decreased levels of the proform of the intrinsic initiator caspase-9 (47 kDa) were seen in A-375 in response to L-779,450 þ TRAIL but not in A-375-TS. Thus, initiator caspases could not (fully) explain enhanced apoptosis (Figure 3a). On the other hand, processing of the main effector caspase-3 was significantly enhanced by L-779,450/TRAIL, seen by its
Figure 2. Sensitization of melanoma cells for tumor necrosis factor–related apoptosis-inducing ligand (TRAIL)-induced apoptosis. (a) Percentages of apoptotic cells (sub-G1) were determined in human melanoma cell lines at 24 hours in response to L-779,450 (10–40 mM) and TRAIL. Examples are shown in insets. (b) Cytotoxicity (lactate dehydrogenase (LDH) release) was determined in parallel. Values of non-treated cells are set to 1. (c) Photographs (magnification 1:400) after bisbenzimide (Hoechst 33258) staining: TRAIL-sensitive A-375 and TRAIL-resistant A-375-TS cells treated for 24 hours with 10 mM L-779,450/TRAIL. White arrows show examples of fragmented or condensed nuclei. (d) Quantitative evaluation of cells with fragmented nuclei and/or condensed chromatin. (e) MeWo cells were treated also for 48 and 72 hours (percentages of sub-G1 cells). (f) Growth curves (real time) of A-375 and A-375-TS treated with TRAIL, L-779,450 (5 mM), or the combination were compared with non-treated controls. (g) Percentages of apoptotic cells (sub-G1) were determined in response to 20 mM U0126±TRAIL. (h) Cell cycle analyses of A-375 and MeWo treated with U0126 (20 mM, 24 hours, open graphs), as compared with dimethyl sulfoxide (DMSO) controls (filled graphs). (i) Growth curves of A-375 treated with U0126 (20 mM) and/or TRAIL (means and SDs of triplicate determinations). (j) Percentages of apoptotic cells (subG1) were determined in response to PLX4032 (3, 10, and 40 mM)±TRAIL. (k) Cell cycle analyses of A-375, A-375-TS, and Mel-HO treated with PLX4032 (10 mM, 2 hours, open graphs), as compared with DMSO controls (filled graphs). Always, two to three independent experiments revealed highly comparable results. Statistical significance (*Po0.05) is indicated, when comparing L-779,450/TRAIL treatment with TRAIL alone. HOECHST/bisbenzimide staining.
www.jidonline.org
433
A Berger et al. RAF in TRAIL Resistance
cleavage products of 15, 17, and 20 kDa (Figure 3a). The decisive role of caspases was further demonstrated by the pancaspase inhibitor Q-VD-OPh, which completely abrogated apoptosis induction by L-779,450/TRAIL (Figure 3b). Lacking enhancement of caspase-8 and Bid processing suggested a role of Smac, which may antagonize cIAPs and thus activate caspase-3. Confirming the activation of mitochondrial apoptosis pathways, strong decrease of the mitochondrial membrane potential Dcm was seen at 24 hours in A-375 (490%) and in A-375-TS (50%) in response to L-779,450/TRAIL. In comparison, L-779,450 alone remained without effect on Dcm, and a response to TRAIL was seen in only 16% of A-375 (Figure 4a). Loss of Dcm appeared as in parallel to apoptosis induction, as not seen at 2 and 4 hours (data not shown). Finally proving the activation of mitochondrial apoptosis pathways, both A-375 and A-375-TS revealed strong release of mitochondrial apoptogenic factors (cytochrome c, AIF, and Smac) upon L-779,450/TRAIL treatment. This effect was already seen at 3 hours, before any apoptosis was induced (Figure 4b) and was still seen at 16 hours (data not shown). Mitochondrial release thus appeared as an early and initial effect. Smac represented a suitable candidate for overcoming the apoptosis blockade through cIAPs in melanoma cells. In agreement, siRNA-mediated Smac knockdown decreased apoptosis by L-779,450/TRAIL in A-375 by 60% (Figure 4c).
Induced Bim expression and autophagy by L-779,450
Decisive roles of Bcl-2 and Bax
DISCUSSION MAPK pathways initiated by Ras/RAF have decisive roles in melanoma (Davies et al., 2002; Jin et al., 2012; Mann et al., 2012). Thus, selective inhibitors for mutated BRAF as vemurafenib and dabrafenib have been approved or are in clinical trials (Flaherty et al., 2010; Chapman et al., 2011; Hauschild et al., 2012; Long et al., 2012; Sosman et al., 2012). The downstream kinase MEK represents an additional promising target, and combinations of both kinase inhibitors are evaluated (Eggermont and Robert, 2011; Flaherty et al., 2012). For targeting the MAPK pathway at an early step, we used here L-779,450, which was described as inhibitor for A-, B-, and CRAF (Shelton et al., 2003). As CRAF activation upon BRAF inhibition is discussed as a cause of resistance and melanoma relapse (Heidorn et al., 2010; Maurer et al., 2011; Sullivan and Flaherty, 2012), simultaneous inhibition of both RAF isoforms may be considered as a supportive strategy. Also other MAPKs, as p38 may be targeted by L-779,450, which may allow a comprehensive antitumor approach (Shelton et al., 2003). Efficient inhibition of the MAPK pathways by L-779,450 was seen in BRAF(V600E)-mutated melanoma cells by complete abrogation of ERK phosphorylation. Comparably high pERK levels were also seen in BRAF-WT cells, which were however less affected by L-779,450. Thus, ERK phosphorylation in these cells may depend on alternative pathways, e.g., as suggested for protein kinase Ca (Rucci et al., 2005). Reduced cell growth in response to L-779,450 was reported in hematopoietic cells, which appeared as related to ARAF and CRAF in these cells (Shelton et al., 2003). Similarly,
The significance of mitochondrial apoptosis pathways was further underscored by decisive roles of Bcl-2 and Bax. Thus, Bcl-2 stable overexpression in A-375 (A-375-Bcl-2) completely prevented apoptosis induction by L-779,450/TRAIL (20 ng ml 1; Figure 5a) and also abolished loss of Dcm (Figure 5b). Because of preferential binding of Bcl-2 to Bax, this suggested a particular role of the Bax pathway. The roles of Bax and Bak were distinguished in a BRAF-WT, K-Ras-mutated HCT-116 colon carcinoma cell model consisting of Bax þ Bak þ parental cells, Bax knockout, Bak knockdown, and double knockdown cells. Comparable apoptosis induction was seen at 24 hours in the parental and the Bak knockdown cells, whereas apoptosis was completely abrogated by Bax knockout (Figure 5c). Bax dependency was also proven in A-375 melanoma cells by siRNA strategies for Bax and Bak, respectively. Again, only the Bax knockdown resulted in resistance to TRAIL as well as to L-779,450/TRAIL, whereas Bak knockdown remained without effect (Figure 5d). The critical role of Bax was further investigated by N-terminus–specific antibodies indicative for Bax activation. Of note, significant Bax activation was seen in A-375 and A-375-TS already in response to L-779,450 alone, which itself did not induce apoptosis. This again appeared as an initial effect already at 2 hours. In contrast, TRAIL treatment remained without effect on Bax activation, and no activation was seen for Bak, as determined by a Bak-NT antibody (Figure 5e). Thus, Bax activation through L-779,450 did not trigger apoptosis by itself but enhanced TRAIL sensitivity of melanoma cells. 434
Journal of Investigative Dermatology (2014), Volume 134
For further understanding the enhancement of apoptosis, the expression of multiple apoptosis regulators was investigated in Mel-HO, A-375, and A-375-TS at 16 hours of treatment. No significant changes in response to L-779,450 were obtained by western blotting for Bcl-2, Mcl-1, Bax, Bak, survivin, or XIAP or by flow cytometry for TRAIL receptors DR4 and DR5 (data not shown). However, significant upregulation was found for the BH3-only protein BimEL, which may contribute to induction of mitochondrial, proapoptotic pathways (Figure 6a). TRAIL alone or in combination with L-779,450 did not affect Bim expression (Figure 6b). The significant role of Bim for L-779,450/TRAIL-mediated apoptosis was shown again by siRNA-mediated Bim knockdown in A-375, which abolished Bim upregulation and decreased apoptosis by 40% (Figure 6c). In course of L-779,450 treatment, responsive melanoma cells revealed consistent morphological changes, as large vacuoles strongly indicative of autophagy (Figure 6d). Again, these changes were not seen in MeWo and SK-Mel-13. Further substantiating autophagy, the marker light chain 3-II (LC3-II) was significantly upregulated upon treatment with L-779,450 (Figure 6e). Thus, apoptosis sensitization for TRAIL by activation of proapoptotic mitochondrial pathways via Bim and Bax as well as induced autophagy appeared as characteristic for the melanoma cell response to RAF inhibition.
A Berger et al. RAF in TRAIL Resistance
A-375-TS
57 41 43
Caspase-8 Bid (proform)
22
Caspase-9 (proform)
47
L-779,450
TRAIL
Control
kDa
A-375
A-375 TS Δm
35 20 17 15
Caspase-3
GAPDH –
+
–
0
+
+
–
0
A-375
20
30 Apoptosis (%)
–
10
+
Q-VD-OPh
TRAIL L-779
* 0
– + – + 0 10
Cyto
Mito
kDa
Cyt c
15
Smac
21
AIF
63
VDAC
31 37
GAPDH
10
TRAIL
–
L-779
–
Q-VD-OPh
* 0
A-375-TS
Cyto
A-375-TS
*
– + – + 0 10
A-375
10
20 10
Mito
– + – + 0 10
– + – + 0 10
Figure 3. Caspase involvement. (a) Processing of caspase-8, -9, and -3 as well as of Bid was determined by western blotting in A-375- and A-375-TS-treated for 16 hours with tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) (20 ng ml 1) and/or L-779,450 (10 mM). Cleavage products of caspase-3 and -8 are marked ( ). (b) Percentages of apoptotic cells (sub-G1) were determined after treatment (24 hours). In addition, the pancaspase inhibitor Q-VD-OPh (10 mM, 1 hour pretreatment, dark gray) was given. Statistical significance of abrogated apoptosis by Q-VD-OPh is indicated (*Po0.05). (a, b) At least two independent experiments revealed comparable results.
melanoma cells responded with reduced cell growth to PLX4720 (Tsai et al., 2008), and antiproliferative effects as well as G1 arrest were also seen here by L-779,450. These antiproliferative effects were not restricted to BRAF-mutated melanoma cells; however, there was a correlation between G1 arrest induced by L-779,450 and its ability to enhance TRAIL-induced apoptosis. TRAIL represents a promising antitumor strategy, due to selective apoptosis induction in tumor cells (Walczak et al., 1999; Ashkenazi, 2002), but the antitumor activity of TRAIL receptor agonists remained only limited in clinical trials (Herbst et al., 2010; Younes et al., 2010; Soria et al., 2011). Non-sufficient efficacy was attributed to inducible resistance, also seen in melanoma cells (Zhang et al., 2006; Kurbanov et al., 2007). Overcoming TRAIL resistance thus represents a promising therapeutic approach, and different strategies were investigated in melanoma cells as signaling inhibitors, chemotherapy, and UV irradiation (Ivanov et al., 2008; Berger et al., 2011; Hornle et al., 2011; Quast et al., 2012).
30 Apoptosis (%)
TRAIL L-779
37
L-779,450 + TRAIL
Counts
A-375
–
+ 0
–
–
+ 10
+ 0
–
+ 10
–
*
Scr
kDa
si Smac 20
21
Smac
37
GAPDH 10
0
Scr
C
–
TRAIL
L-779
si Smac
TRAIL/ L-779
Figure 4. Activation of mitochondrial (Mito) apoptosis pathways. (a) Mitochondrial membrane potential (Dcm) was determined in A-375 and A-375-TS treated for 24 hours with tumor necrosis factor–related apoptosisinducing ligand (TRAIL) (20 ng ml 1) and/or L-779,450 (10 mM). Treated cells (open graphs) were compared with dimethyl sulfoxide (DMSO) controls (gray). (b) Cytosolic extracts (Cyto) of TRAIL-sensitive A-375 and TRAIL-resistant A-375-TS were investigated by western blotting, after treatment for 3 hours with TRAIL±L-779,450 (10 mM). Mitochondrial extracts (Mito) served as controls, and analysis of the mitochondrial protein voltage-dependent anion channels (VDAC) ruled out contaminations of cytosolic extracts with mitochondria. Equal loading was proven by glyceraldehyde dehydrogenase (GAPDH). (c) Percentages of apoptotic cells (sub-G1) were determined in A-375 in response to 10 mM L-779,450±TRAIL (24 hours). Cells with small interfering (si)RNAmediated Smac knockdown (dark bars) were compared with scramble (Scr)-siRNA-transfected controls (light gray; means and SDs of triplicate values). Smac downregulation is shown by western blotting (inset; 24 hours, GAPDH as control). Statistical significance of abrogated apoptosis by Smac knockdown is indicated (*Po0.05). (a–c) At least two independent experiments revealed comparable results. AIF, apoptosis-inducing factor; Cyt c, cytochrome c; Dcm, mitochondrial membrane potential.
Although L-779,450 hardly induced apoptosis by itself, it efficiently enhanced TRAIL-induced apoptosis in eight of ten melanoma cell lines, suggesting a link between RAF activation and TRAIL resistance in melanoma cells. The relation between active MAPK pathways and TRAIL resistance had also been reported previously by using an inhibitor for the downstream MAPK MEK (Zhang et al., 2003). Activation of MAPK www.jidonline.org
435
A Berger et al. RAF in TRAIL Resistance
GAPDH
TRAIL C lgG
C T/L
kDa 26
Bcl-2
37
A-375 Mock Bcl-2 10
A-375 BCL-2
* – + A-375-mock
L-779
*
L-779 TRAIL
*
*
– + A-375-Bcl-2 Δψm
50
*
Counts
20
0 L-779
Bax-NT
A-375-mock
TRAIL – +
Counts
Apoptosis (%)
30
TRAIL – +
kDa Bax
25
Bak
28
A-375
A-375-TS
Bak-NT
30 37
GAPDH 20
Bax – Bax Bak Bak –
10
–
HCT-
Bax/Bax
+
–
+
–
Bax / –
C lgG
– –
*
0 L-779
TRAIL
*
+
–
– / Bak
L-779
+
Counts
Apoptosis (%)
40
–/– kDa
Apoptosis (%)
30
TRAIL – +
20
Bax
25
Bak
28
0 L-779
37
GAPDH Scr
10
L-779 TRAIL
*
sisiBax Bax A-375
– + Scramble
–
+ si-Bak
A-375-TS
– + si-Bax
Figure 5. Dependency and activation of Bax. (a, c, d) Percentages of apoptotic cells (sub-G1) were determined in A-375 cells stably transfected with Bcl-2 (A-375-Bcl-2) or mock plasmid (A-375-mock), parental HCT-116 colon carcinoma cells (Bax þ , Bak þ ), and subclones with knockdown for Bax and/or Bak, as well as in A-375 cells with small interfering RNA (siRNA)-mediated knockdown for either Bax or Bak. Treatment was for 24 hours, and tumor necrosis factor–related apoptosis-inducing ligand (TRAIL)-treated cells (dark bars) were compared with cells without TRAIL (light gray). (a, c) L-779,450 was applied at 5 mM, but comparable results were also obtained for 10 mM (data not shown). (d) L-779,450 was applied at 10 mM. Means and SDs of representative experiments (triplicate values) are shown. Expression of Bcl-2, Bax, and Bak is shown in respective insets (24 hours, western blotting, glyceraldehyde dehydrogenase (GAPDH) as loading control). Statistical significance of differences is indicated (*Po0.05), when compared with parental/mock cells. (b) Mitochondrial membrane potential (Dcm) was determined in A-375-Bcl-2 and A-375-mock. L-779,450 (5 mM)/TRAIL treatment (open graphs) was compared with dimethyl sulfoxide (DMSO) controls (gray). (e) A-375 and A-375-TS cells were treated for 2 hours with L-779,450 (10 mM) and/or TRAIL and were analyzed for Bax/Bak conformational changes by flow cytometry (Bax-NT/Bak-NT antibodies). Treated cells (bold lines) were compared with non-treated controls (C, gray) and immunoglobulin G (IgG)–stained cells (thin lines). (a–e) At least two independent experiments revealed comparable results. Scr, scramble.
pathways and relations to TRAIL resistance were also seen in colon carcinoma (Oikonomou et al., 2011), and in agreement we found enhancement of TRAIL-induced apoptosis in BRAFWT- and K-Ras-mutated HCT-116 colon carcinoma cells. As also BRAF-WT and N-Ras-mutated melanoma cell lines were sensitized for TRAIL by L-779,450, pan-RAF inhibition also 436
Journal of Investigative Dermatology (2014), Volume 134
proved helpful when the pathway was activated at an upstream step. Enhanced activation of effector caspase-3, whereas activation of initiator caspase-8 and -9 was not further enhanced by L-779,450, indicated the role of mitochondrial apoptosis pathways and Smac release. Smac can activate caspase-3
A Berger et al. RAF in TRAIL Resistance
via preventing the blockage of XIAP (Chai et al., 2000). Thus, caspase-3 was processed to its intermediate 20 kDa cleavage product in response to TRAIL, whereas full caspase-3 processing to its mature 15 and 17 kDa cleavage products was particularly enabled upon combination with L-779,450. The significance of the Smac/XIAP rheostat in melanoma apoptosis resistance was also suggested in previous studies (Zhang et al., 2003; Hornle et al., 2011; Quast et al., 2012). Thus, mitochondrial apoptosis pathways were activated by
A-375
Mel-HO
A-375-TS
kDa
BimEL
24
GAP DH L-779
37 +
–
–
+
–
+
A-375-TS
A-375
kDa
BimEL
24
GAP DH TRAIL L-779
Apoptosis (%)
25 20
37 – –
+ –
– +
+ +
– –
+ –
+ +
si si Scr Bim Scr Bim
*
A-375 Scr siBim
– +
BimEL GAPDH
15
L-779
10
–
–
+
+
5 0
TRAIL L-779 TRAIL/ L-779
C
Control
L-779,450
A-375
A-375 TS
A-375
kDa
A-375-TS
LC3-II
16
GAPDH TRAIL L-779
37 – –
+ –
– +
+ +
– –
+ –
– +
+ +
L-779,450/TRAIL already after 3 hours, as seen by the release of apoptogenic mitochondrial factors. A role of mitochondrial pathways had also been seen in melanoma cells when combining other signaling inhibitors with TRAIL (Zhang et al., 2003; Berger et al., 2011; Quast et al., 2012). XIAP is considered as a major anti-apoptotic factor in cancer cells, which prevents caspase-3 activity (Mannhold et al., 2010; Fulda and Vucic, 2012), and its predominant role was demonstrated in melanoma cells (Hornle et al., 2011; Quast et al., 2012). It may therefore also explain enhanced apoptosis by L-779,450/TRAIL. In agreement, siRNA-mediated knockdown largely abolished induced apoptosis. Suppression of apoptosis by Smac knockdown was also seen in melanoma cells, when TRAIL was combined with UVB or an ion channel inhibitor (Hornle et al., 2011; Quast et al., 2012). Bcl-2 proteins, which control mitochondrial pathways, critically affected apoptosis induction by L-779,450/TRAIL. Thus, apoptosis was abrogated in melanoma cells by Bcl-2 overexpression or Bax knockdown. The decisive role of Bax activation was proven by Bax N-terminus-specific antibodies, whereas the alternative pathway via Bak was not affected. Interestingly, Bax was activated by L-779,450 treatment alone already after 2 hours. As L-779,450 did not induce apoptosis by itself, it only opened a gate for TRAIL sensitization, and the switch appeared to be the Bax activation. Apoptosis regulators may be controlled by MAPK pathways, as upregulation of Mcl-1 and survivin or downregulation of Bad and Bim (Ai et al., 2006; McCubrey et al., 2007; Cartlidge et al., 2008). Whereas no significant changes were obtained for other apoptosis regulators, the proapoptotic BH3-only protein BimEL was significantly upregulated in melanoma cells in response to L-779,450. Upregulation of Bim was also reported in response to the selective BRAF inhibitor PLX4720 (Wroblewski et al., 2013) and the MEK inhibitor U0126 (Wang et al., 2007). Bim isoforms represent efficient inductors of apoptosis through antagonizing all prosurvival Bcl-2 proteins (Chen et al., 2005). The relation of Bim and MAPKs is based on ERK-mediated Bim phosphorylation, which triggers its proteasomal degradation (Cartlidge et al., 2008). The critical role of Bim in sensitization of melanoma
Figure 6. Induction of Bim and autophagy. (a, b) Expression of BimEL was determined by western blotting in Mel-HO, A-375, and A-375-TS melanoma cells treated with 10 mM L-779,450 and in A-375 and A-375-TS treated with L-779,450±tumor necrosis factor–related apoptosis-inducing ligand (TRAIL). Glyceraldehyde dehydrogenase (GAPDH) served as loading control. (c) Percentages of apoptotic cells (sub-G1) were determined in A-375 cells with small interfering RNA (siRNA)-mediated knockdown for Bim (dark bars), as compared with scramble (Scr)-siRNA-transfected controls (light gray). Treatments: 10 mM L-779,450±TRAIL for 24 hours. Means and SDs of representative experiments (triplicate values) are shown. Statistical significance is indicated (*Po0.05), when comparing Bim and scramble siRNA. (Inset) Bim expression is shown by western blotting (24 hours), with GAPDH as loading control. (d) Cell photographs (magnification 1:200) of TRAIL-sensitive A-375 and TRAIL-resistant A-375-TS treated with L-779,450 (10 mM) are shown. (e) Expression of light chain 3-II (LC3-II) was determined by western blotting in A-375 and A-375-TS treated with TRAIL (20 ng ml 1) and/or L-779,450 (10 mM). (a–e) At least two independent experiments revealed comparable results.
www.jidonline.org
437
A Berger et al. RAF in TRAIL Resistance
cells for TRAIL-induced apoptosis by L-779,450 was demonstrated here by its siRNA-mediated knockdown. Also, induced autophagy was seen upon L-779,450 treatment. Autophagy mainly represents a survival mechanism, which may enable tumor cells to outlive limiting growth conditions (Kondo et al., 2005; Levine, 2006). Thus, autophagy induced by MAPK inhibition may be seen as a response of the cells to survive the limitation caused by RAF inhibition. Enhanced autophagy in melanoma cells was recently also reported in response to selective BRAF inhibitors (Kim et al., 2012). Autophagy may thus limit an antitumor activity (Gump and Thorburn, 2011), but a possible autophagy-mediated survival was crossed here by apoptosis induction through TRAIL. In conclusion, inhibition of cell proliferation and sensitization for TRAIL-induced apoptosis are suggested here for explaining the antitumor activity of RAF inhibitors in melanoma. As TRAIL is expressed by cytotoxic T cells (Brincks et al., 2011), enhancement of TRAIL sensitivity may further support an antitumor immune response. Moreover, TRAIL receptor agonists, which have been seen in clinical trials, in combination with RAF inhibitors may be considered for melanoma therapy. MATERIALS AND METHODS Cell culture Experiments were performed in TRAIL-sensitive (A-375, Mel-HO, SK-Mel-28, SK-Mel-13, and SK-Mel-147) and in permanent resistant (Mel-2a, MeWo, and SK-Mel-103) human melanoma cell lines (Kurbanov et al., 2005). Mel-HO and A-375 were derived from primary tumors, whereas Mel-2a, SK-Mel-13, MeWo, SK-Mel-28, SK-Mel-147, and SK-Mel-103 were from metastasis. Sublines with acquired TRAIL resistance (Mel-HO-TS and A-375-TS) were derived from selection with 100 ng ml 1 TRAIL (Kurbanov et al., 2007). TRAIL resistance was regularly controlled in parallel to all experiments. Other A-375 subclones had been stably transfected with a pIRES-Bcl-2 plasmid (A-375-Bcl-2) or pIRES mock plasmid (A-375-mock), as described previously (Raisova et al., 2001). Parental BRAF-WT, K-Ras-mutated HCT-116 colon carcinoma cells were derived from ATCC (Maryland, MD). The HCT-116 Bax knockout, Bak knockdown, and Bax/Bak double knockdown cells were kindly provided by B Vogelstein (John Hopkins Cancer Center, Baltimore, MD) and had been described previously (Gillissen et al., 2010). Cells were cultivated at 37 1C, 5% CO2 in DMEM (4.5 g l 1 glucose; Gibco, Invitrogen, Karlsruhe, Germany) with 10% fetal calf serum (FCS) and antibiotics (Biochrom, Berlin, Germany). TRAILselected cells were continuously kept with 5 ng ml 1 TRAIL until 24 hours before treatment. Cells were plated in 6-, 24-, or 96-well plates with 2 105, 5 104, and 5 103 cells, respectively, and treatment started after 24 hours. For induction of apoptosis, TRAIL (Alexis, Gruenberg, Germany; ALX-201-073-C020; 20 ng ml 1), the pan-RAF inhibitor L-779,450 (Merck, Darmstadt, Germany; 0.1–50 mM), the MEK inhibitor U0126 (Sigma-Aldrich, Taufkirchen, Germany; 20 mM), and the selective BRAF(V600E) inhibitor vemurafenib/PLX4032 (Selleck Chemicals, Houston, TX) were used. For caspase inhibition, cells were preincubated for 1 hour with 10 mM of Q-VD-OPh (MP Biomedicals, Solon, OH). 438
Journal of Investigative Dermatology (2014), Volume 134
Cell proliferation, apoptosis, and cytotoxicity For continuous monitoring cell growth, the xCELLigence system (Roche Diagnostics, Penzberg, Germany) was applied. Relative cell indices correspond to attached cell numbers. Cell cycle analyses were performed for quantification of apoptosis and cell cycle arrest (Riccardi and Nicoletti, 2006). Cells harvested by trypsinization were stained for 1 hour with propidium iodide (Sigma-Aldrich; 200 mg ml 1), and sub-G1 fractions, corresponding to cells with fragmented DNA, were quantified by flow cytometry (FACS Calibur; BD Bioscience, Bedford, MA). Cytotoxicity was determined according to the release of lactate dehydrogenase by damaged cells into the culture supernatants. Values of non-treated cells were set to 1, and data were calculated as relative values of the non-treated controls (Cytotoxicity detection assay; Roche Diagnostics). Mitochondrial membrane potential was determined with the fluorescent dye tetramethylrhodamine methyl ester perchlorate Sigma-Aldrich; 1 mM (TMRM þ ; Sigma-Aldrich; 1 mM). Cells harvested by trypsinization were stained for 15 min at 37 1C and analyzed by flow cytometry. For identification of chromatin condensation and nuclear fragmentation in course of apoptosis, cells were harvested by trypsinization, centrifuged on cytospins, and fixed for 30 minutes in 4% formaldehyde. Cytospins were stained with bisbenzimide (Hoechst-33258; Sigma-Aldrich; 1 mg ml 1, 30 minutes) and examined by fluorescence microscopy. Apoptotic cells were identified by fragmented nuclei or by bright blue-stained nuclei with condensed chromatin.
Assays for Bax/Bak activation For analysis of Bax and Bak conformational changes related to their activation, primary antibodies specific for the N-terminal domains of Bax or Bak were applied in flow cytometry (Bax-NT, 1:100 (Upstate, Lake Placid, NY; no. 06-499); Bak-NT (Merck, Darmstadt, Germany), AM04, 1:10). The N-terminal domains become accessible in course of Bax/Bak activation (Gillissen et al., 2007; Martinou and Youle, 2011). Cells (105) were harvested by trypsinization and fixed for 30 minutes with 4% paraformaldehyde in phosphate-buffered saline. Cells were then incubated with the respective antibodies for 1 hour at 4 1C in phosphate-buffered saline/1% FCS, which also contained 0.1% saponin for cell permeabilization. Cells were subsequently incubated for 1 hour at 4 1C in the dark with the secondary antibody goat anti-rabbit IgG (H þ L)-FITC (Jackson Immuno Research, West Grove, FL). After washing and resuspension in phosphate-buffered saline/1% FCS, cells were immediately measured by flow cytometry.
Cell transfection with siRNA Transient cell transfection was performed in 6-well plates at 24 hours after seeding (70% confluence). Treatment with L-779,450/TRAIL followed after another 24 hours. Amounts of 20 pmol siRNA and 4 ml TurboFect (Fermentas, St Leon-Rot, Germany) were used per well. The siRNAs for Smac (sc-36505), Bax (sc-29212), Bak (sc-29786), Bim (sc-29802), and the scrambled control (sc-37007) were derived from Santa Cruz Biotechnology (Santa Cruz, CA).
Protein analysis For western blotting, total protein extracts were obtained by cell lysis in 150 mM NaCl, 1 mM EDTA, 2 mM phenylmethyl sulfonyl, 1 mM leupeptin, 1 mM pepstatin, 0.5% SDS, 0.5% NP-40, and 10 mM Tris-HCl (pH 7.5). Cytosolic and mitochondrial cell fractions were prepared by a
A Berger et al. RAF in TRAIL Resistance
kit of PromoKine (Heidelberg, Germany). Western blotting on nitrocellulose membranes was described previously (Eberle et al., 2003). Primary antibodies obtained were as follows: cleaved caspase-3 (3664, rabbit, 1:5,000), caspase-3 proform (9662, rabbit, 1:1,000), caspase-8 (9746, mouse, 1:1,000), caspase-9 (9505, rabbit, 1:1,000), ERK (4695, rabbit, 1:2,000), pERK (9101, rabbit, 1:2,000), and XIAP (2042, rabbit, 1:1,000) from Cell Signaling; Bcl-2 (sc-783, rabbit; 1:200), Bax (sc-493, rabbit, 1:200), Bak (sc-893, rabbit, 1:500), AIF (sc-9416, goat, 1:1,000), Smac (sc-56230, mouse, 1:1,000), Bim (sc11425, rabbit, 1:400), light chain 3 (sc-292354, rabbit, 1:500), Bid (sc-2002, rabbit, 1:1,000), glyceraldehyde dehydrogenase (GAPDH) (sc-3233, mouse, 1:1,000), and survivin (sc-17779, mouse, 1:500) from Santa Cruz Biotechnology. The antibodies used further were: cytochrome c (no. 556433, mouse, 1:1,000; BD Biosciences, Heidelberg, Germany) and VDAC (voltage-dependent anion channels; antiporin 31HL, 529536, mouse, 1:500; Calbiochem, Darmstadt, Germany). The secondary antibodies used were as follows: peroxidaselabeled goat anti-rabbit, goat anti-mouse, and rabbit anti-goat (Dako, Hamburg, Germany; 1:5,000).
Statistical analyses Assays consisted of triplicates, and at least two independent experiments were performed. Mean values and SDs were calculated either by enclosing all individual values of the independent experiments (at least six values) or a representative experiment is shown (triplicate values). Statistical significance was proven by Student’s t-test in case of normal distribution, and P-values of o0.05 were considered as statistically significant. CONFLICT OF INTEREST The authors state no conflict of interest.
ACKNOWLEDGMENTS The study was supported by the German Cancer Aid (Deutsche Krebshilfe), Melanomverbund, 10-8008, TP7.
REFERENCES Ai ZH, Yin LH, Zhou XR et al. (2006) Inhibition of survivin reduces cell proliferation and induces apoptosis in human endometrial cancer. Cancer 4:746–56 Ashkenazi A (2002) Targeting death and decoy receptors of the tumournecrosis factor superfamily. Nat Rev Cancer 6:420–30 Berger A, Quast SA, Plotz M et al. (2011) Sensitization of melanoma cells for death ligand-induced apoptosis by an indirubin derivative—enhancement of both extrinsic and intrinsic apoptosis pathways. Biochem Pharmacol 1:71–81 Brincks EL, Gurung P, Langlois RA et al. (2011) The magnitude of the T cell response to a clinically significant dose of influenza virus is regulated by TRAIL. J Immunol 9:4581–8 Cartlidge RA, Thomas GR, Cagnol S et al. (2008) Oncogenic BRAF(V600E) inhibits BIM expression to promote melanoma cell survival. Pigment Cell Melanoma Res 5:534–44 Chai JJ, Du CY, Wu JW et al. (2000) Structural and biochemical basis of apoptotic activation by Smac/DIABLO. Nature 6798:855–62 Chapman PB, Hauschild A, Robert C et al. (2011) Improved survival with Vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 26:2507–16 Chen L, Willis SN, Wei A et al. (2005) Differential targeting of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apoptotic function. Mol Cell 3:393–403 Chipuk JE, Moldoveanu T, Llambi F et al. (2010) The BCL-2 family reunion. Mol Cell 3:299–310
Davies H, Bignell GR, Cox C et al. (2002) Mutations of the BRAF gene in human cancer. Nature 6892:949–54 Eberle J, Fecker LF, Hossini AM et al. (2003) CD95/Fas signaling in human melanoma cells: conditional expression of CD95L/FasL overcomes the intrinsic apoptosis resistance of malignant melanoma and inhibits growth and progression of human melanoma xenotransplants. Oncogene 57: 9131–41 Eberle J, Kurbanov BM, Hossini AM et al. (2007) Overcoming apoptosis deficiency of melanoma—hope for new therapeutic approaches. Drug Resist Updates 6:218–34 Eggermont AMM, Robert C (2011) New drugs in melanoma: It’s a whole new world. Eur J Cancer 14:2150–7 Fischer U, Janicke RU, Schulze-Osthoff K (2003) Many cuts to ruin: a comprehensive update of caspase substrates. Cell Death Differ 1:76–100 Flaherty KT, Infante JR, Daud A et al. (2012) Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N Engl J Med 18:1694–703 Flaherty KT, Puzanov I, Kim KB et al. (2010) Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med 9:809–19 Fulda S, Vucic D (2012) Targeting IAP proteins for therapeutic intervention in cancer. Nat Rev Drug Disc 2:109–24 Garbe C, Leiter U (2009) Melanoma epidemiology and trends. Clin Dermatol 1:3–9 Gillissen B, Essmann F, Hemmati PG et al. (2007) Mcl-1 determines the Bax dependency of Nbk/Bik-induced apoptosis. J Cell Biol 4:701–15 Gillissen B, Wendt J, Richter A et al. (2010) Endogenous Bak inhibitors Mcl-1 and Bcl-x(L): differential impact on TRAIL resistance in Bax-deficient carcinoma. J Cell Biol 6:851–62 Gray-Schopfer VC, Dias SD, Marais R (2005) The role of B-RAF in melanoma. Cancer Metast Rev 1:165–83 Gump JM, Thorburn A (2011) Autophagy and apoptosis: what is the connection? Trends Cell Biol 7:387–92 Hauschild A, Grob JJ, Demidov LV et al. (2012) Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet 9839:358–65 Heidorn SJ, Milagre C, Whittaker S et al. (2010) Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell 2:209–21 Herbst RS, Eckhardt SG, Kurzrock R et al. (2010) Phase I dose-escalation study of recombinant human Apo2L/TRAIL, a dual proapoptotic receptor agonist, in patients with advanced cancer. J Clin Oncol 17: 2839–46 Hornle M, Peters N, Thayaparasingham B et al. (2011) Caspase-3 cleaves XIAP in a positive feedback loop to sensitize melanoma cells to TRAIL-induced apoptosis. Oncogene 5:575–87 Inamdar GS, Madhunapantula SV, Robertson GP (2010) Targeting the MAPK pathway in melanoma: Why some approaches succeed and other fail. Biochem Pharmacol 5:624–37 Ivanov VN, Partridge MA, Johnson GE et al. (2008) Resveratrol sensitizes melanomas to TRAIL through modulation of antiapoptotic gene expression. Exp Cell Res 5:1163–76 Jin SA, Chun SM, Choi YD et al. (2012) BRAF mutations and KIT aberrations and their clinicopathological correlation in 202 Korean melanomas. J Invest Dermatol 133:579–82 Kim YK, Ahn SK, Lee M (2012) Differential sensitivity of melanoma cell lines with differing B-Raf mutational status to the new oncogenic B-Raf kinase inhibitor UI-152. Cancer Lett 2:215–24 Kondo Y, Kanzawa T, Sawaya R et al. (2005) The role of autophagy in cancer development and response to therapy. Nat Rev Cancer 9: 726–34 Krammer PH, Arnold R, Lavrik IN (2007) Life and death in peripheral T cells. Nat Rev Immunol 7:532–42 Kurbanov BM, Fecker LF, Geilen CC et al. (2007) Resistance of melanoma cells to TRAIL does not result from upregulation of antiapoptotic proteins by NF-kappa B but is related to downregulation of initiator caspases and DR4. Oncogene 23:3364–77
www.jidonline.org
439
A Berger et al. RAF in TRAIL Resistance
Kurbanov BM, Geilen CC, Fecker LF et al. (2005) Efficient TRAIL-R1/DR4mediated apoptosis in melanoma cells by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). J Invest Dermatol 5:1010–9 Levine B (2006) Unraveling the role of autophagy in cancer. Autophagy 2:65–6 Li HL, Zhu H, Xu CJ et al. (1998) Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis. Cell 4:491–501 Long GV, Trefzer U, Davies MA et al. (2012) Dabrafenib in patients with Val600Glu or Val600Lys BRAF-mutant melanoma metastatic to the brain (BREAK-MB): a multicentre, open-label, phase 2 trial. Lancet Oncol 13:1087–95
Shelton JG, Moye PW, Steelman LS et al. (2003) Differential effects of kinase cascade inhibitors on neoplastic and cytokine-mediated cell proliferation. Leukemia 9:1765–82 Soria JC, Mark Z, Zatloukal P et al. (2011) Randomized phase II study of dulanermin in combination with paclitaxel, carboplatin, and bevacizumab in advanced non-small-cell lung cancer. J Clin Oncol 33:4442–51 Sosman JA, Kim KB, Schuchter L et al. (2012) Survival in BRAF V600-mutant advanced melanoma treated with vemurafenib. N Engl J Med 8:707–14
Mann GJ, Pupo GM, Campain AE et al. (2012) BRAF mutation, NRAS mutation, and the absence of an immune-related expressed gene profile predict poor outcome in patients with Stage III melanoma. J Invest Dermatol 133:509–17
Sullivan RJ, Flaherty K (2012) MAP kinase signaling and inhibition in melanoma. Oncogene 32:2373–9
Mannhold R, Fulda S, Carosati E (2010) IAP antagonists: promising candidates for cancer therapy. Drug Discov Today 5–6:210–9
Tsai J, Lee JT, Wang W et al. (2008) Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity. Proc Natl Acad Sci USA 8:3041–6
Martinou JC, Youle RJ (2011) Mitochondria in apoptosis: Bcl-2 family members and mitochondrial dynamics. Dev Cell 1:92–101 Maurer G, Tarkowski B, Baccarini M (2011) Raf kinases in cancer-roles and therapeutic opportunities. Oncogene 32:3477–88 McCubrey JA, Steelman LS, Chappell WH et al. (2007) Roles of the Raf/MEK/ ERK pathway in cell growth, malignant transformation and drug resistance. Biochim Biophys Acta 8:1263–84 Newsom-Davis T, Prieske S, Walczak H (2009) Is TRAIL the holy grail of cancer therapy? Apoptosis 4:607–23 Oikonomou E, Koc M, Sourkova V et al. (2011) Selective BRAFV600E inhibitor PLX4720, requires TRAIL assistance to overcome oncogenic PIK3CA resistance. PLos One 6:e21632 Quast SA, Berger A, Buttstadt N et al. (2012) General sensitization of melanoma cells for TRAIL-induced apoptosis by the potassium channel inhibitor TRAM-34 depends on release of SMAC. PLoS One 6:e39290 Raisova M, Hossini A, Riebeling C et al. (2001) Apoptosis resistance in human melanoma cells. J Invest Dermatol 1:168 Riccardi C, Nicoletti I (2006) Analysis of apoptosis by propidium iodide staining and flow cytometry. Nat Protoc 3:1458–61 Rucci N, DiGiacinto C, Orru L et al. (2005) A novel protein kinase C alphadependent signal to ERK1/2 activated by alpha(V)beta(3) integrin in
440
osteoclasts and in Chinese hamster ovary (CHO) cells. J Cell Sci 15:3263–75
Journal of Investigative Dermatology (2014), Volume 134
Tait SWG, Green DR (2010) Mitochondria and cell death: outer membrane permeabilization and beyond. Nat Rev Mol Cell Biol 9:621–32
Walczak H, Miller RE, Ariail K et al. (1999) Tumoricidal activity of tumor necrosis factor related apoptosis-inducing ligand in vivo. Nat Med 2: 157–63 Wang YF, Jiang CC, Kiejda KA et al. (2007) Apoptosis induction in human melanoma cells by inhibition of MEK is caspase-independent and mediated by the Bcl-2 family members PUMA, Bim, and Mcl-1. Clin Cancer Res 16:4934–42 Wroblewski D, Mijatov B, Mohana-Kumaran N et al. (2013) The BH3-mimetic ABT-737 sensitizes human melanoma cells to apoptosis induced by selective BRAF inhibitors but does not reverse acquired resistance. Carcinogenesis 2:237–47 Younes A, Vose JM, Zelenetz AD et al. (2010) A phase 1b/2 trial of mapatumumab in patients with relapsed/refractory non-Hodgkin’s lymphoma. Br J Cancer 12:1783–7 Zhang XD, Borrow JM, Zhang XY et al. (2003) Activation of ERK1/2 protects melanoma cells from TRAIL-induced apoptosis by inhibiting Smac/ DIABLO release from mitochondria. Oncogene 19:2869–81 Zhang XD, Wu JJ, Gillespie S et al. (2006) Human melanoma cells selected for resistance to apoptosis by prolonged exposure to tumor necrosis factorrelated apoptosis-inducing ligand are more vulnerable to necrotic cell death induced by cisplatin. Clin Cancer Res 4:1355–64