Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

Biol. Cell (2011) 103, 21–33 (Printed in Great Britain) doi:10.

1042/BC20100094
Research article

PMA up-regulates the transcription


of Axl by AP-1 transcription factor
binding to TRE sequences via the
MAPK cascade in leukaemia cells
Giridhar Mudduluru∗ , Jörg H. Leupold∗ , Philipp Stroebel† and Heike Allgayer∗1
∗ Department of Experimental Surgery Mannheim/Molecular Oncology of Solid Tumors, DKFZ and University of Heidelberg, Mannheim
www.biolcell.org

68167, Germany, and †Institute of Pathology, Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany

Background. Axl is a receptor tyrosine kinase promoting anti-apoptosis, invasion and mitogenesis, and is highly
expressed in different solid cancers. Axl basal transcriptional activity is driven by Sp1/Sp3, and overexpression
of MZF-1 (myeloid zinc-finger 1) induces Axl transcription and gene expression. Furthermore, Axl expression is
epigenetically controlled by CpG hypermethylation; however, little is known about inducible Axl gene expression
and Axl regulation in haematopoetic malignancies.
Results. In the present study, we studied Axl transcriptional regulation under PMA-stimulated conditions in leuk-
aemia cells. Luciferase analysis with sequential 5 -deletion constructs revealed that the − 660/ − 580 region of
the Axl promoter is indispensable for induced promoter activity under PMA stimulation. This region includes AP-1
(activator protein 1)/CREB [CRE (cAMP-response-element)-binding protein] motifs, five times partially overlap-
ping TGCGTG repeats and multiple GT repeats. Mutational, supershift and ChIP (chromatin immunoprecipitation)
analysis determined that AP-1 family members bind to AP-1 motifs and to the 5 × TGCGTG overlapping re-
peats, thus transactivating Axl promoter activity. Furthermore, specific inhibitors of PKC (protein kinase C), ERK1/2
(extracellular-signal-regulated kinase 1/2) and p38 reduced Axl expression. Additionally, mithramycin treatment
abolished constitutive and PMA-induced Axl expression.
Conclusions. Taken together the results of the present study suggest that PMA-induced Axl gene expression in
Biology of the Cell

leukaemia cells is mediated by AP-1 motifs and 5 × TGCGTG repeats within the promoter region − 660/ − 580,
and through the PKC/ERK1/2/AP-1 or PKC/p-38/AP-1 signalling axis.

Introduction
Axl receptor tyrosine kinase belongs to the Tyro3
family, originally identified as a transforming gene
in human leukaemias (Janssen et al., 1991; O’Bryan
1 Towhom correspondence should be addressed (email et al., 1991). The human Axl protein has a molecular
heike.allgayer@umm.de). mass of 140 kDa, and an approximately equal distri-
Key words: activator protein 1 (AP-1), Axl promoter, PMA, receptor tyrosine
kinase. bution of amino acids on either side of the plasma
Abbreviations used: AP-1, activator protein 1; ATF, activating transcription membrane with two immunoglobulin-like domains
factor; ChIP, chromatin immunoprecipitation; CRE, cAMP-response element;
CREB, CRE-binding protein; EGF, epidermal growth factor; EMSA, and two fibronectin-type III domains in the extra-
electrophoretic mobility-shift assay; ERK, extracellular-signal-regulated cellular region, and a distinctive intracellular kinase
kinase; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; JDP,
J-domain-containing protein; JNK, c-Jun N-terminal kinase; MAPK, domain (O’Bryan et al., 1991; Graham et al., 1994;
mitogen-activated protein kinase; MMP, matrix metalloproteinase; MZF-1, Lai et al., 1994). Gas6 is a known ligand for Axl,
myeloid zinc finger 1; NF-κB, nuclear factor κB; PKC, protein kinase C; sAxl,
soluble Axl; TRE, PMA-response element; TTBS, Tris-buffered saline with
and the Gas6–Axl axis is known to transduce anti-
0.1% Tween; uPA, urokinase-type plasminogen activator; uPAR, uPA receptor. apoptotic signals (Demarchi et al., 2001). Axl is not

www.biolcell.org | Volume 103 (1) | Pages 21–33 21


G. Mudduluru and others

only expressed as a transmembrane protein, but is also carcinogenesis (Angel and Karin, 1991; Shaulian and
cleaved at its extracellular domain to produce a sAxl Karin, 2002, Eferl and Wagner, 2003).
(soluble Axl) form of ∼65 kDa. In mouse cells, it Our previous studies (Mudduluru and Allgayer,
has been shown that sAxl generation is mediated by 2008; Mudduluru et al., 2010) have shown that con-
ADAM10 (a disintegrin and metalloproteinase 10) stitutive expression of Axl in solid cancer is driven by
proteolysis, and high levels of sAxl were found as a Sp1/Sp3, and that overexpression of MZF-1 (myeloid
heterocomplex with the ligand Gas6 in serum (Costa zinc finger 1) transactivates Axl transcription and
et al., 1996; Budagian et al., 2005a). Overexpression gene expression. Moreover, Axl and MZF-1 are co-
of Axl can transform fibroblasts, even in the absence of expressed in colorectal cancer tissues. Furthermore,
Gas6 (Burchert et al., 1998) and is associated with in- Axl expression is epigenetically controlled by CpG
vasiveness and metastasis in various cancer types. Axl hypermethylation. Still, the transcriptional regula-
overexpression was also detected in peritoneal meta- tion of Axl in blood cells and under stimulated con-
static nodules of colon cancers, prostate carcinoma, ditions is not clearly known. Therefore, the present
gastric sarcoma, certain types of breast cancers, myel- study was performed with leukaemia cells as a model
oid leukaemia and some other cancer types (Neubauer system, and with PMA as an oncogenic stimulus
et al., 1994; Weiner et al., 1994; Craven et al., 1995; to investigate the essential promoter region of Axl,
Jacob et al., 1999; Meric et al., 2002; Wu et al., 2002; its transcriptional regulation and inducing signalling
Sun et al., 2003). molecules under stimulated conditions.
Signals transmitted through PKC (protein kinase
C) can potentially activate the network of MAPK
(mitogen-activated protein kinase), which plays an Results
important role in regulating the response of cells to PMA (5 nM) induces Axl gene expression
various extracellular stimuli, such as PMA (Rubin- in leukaemia cells
feld and Seger, 2004; Kolch et al., 2005). The MAPK To determine the effect of PMA on Axl expression,
signalling pathways are activated by sequential phos- K562 and HL-60 cells were stimulated with PMA in
phorylation steps, through five pathways mediated by a dose-dependent manner for 24 h. Endogenous Axl
JNK (c-Jun N-terminal kinase), ERK (extracellular- mRNA amounts were screened by quantitative-PCR
signal-regulated kinase) 1/2, p38 MAPK, ERK5 and analysis. Both cell lines showed inducible Axl gene
ERK3/4. Activated MAPK kinase pathways activ- expression in a dose-dependent manner (Figures 1a
ate AP-1 (activator protein 1), which is a heterodi- and 1b). We determined that 5 nM PMA is suffi-
meric complex of structurally and functionally re- cient for stimulation of K562 and HL-60 cells, in
lated members of the Jun (c-Jun, JunB or JunD) which a 2-fold (K562) and 10-fold (HL-60) increase
and Fos (c-Fos, FosB, Fra-1 or Fra-2) family. Ad- in Axl gene expression is shown (Figures 1a and 1b).
ditionally, some members of the ATF (activating Furthermore, 5 nM PMA stimulation significantly
transcription factor) (ATFa, ATF-2 and ATF-3) and induced Axl protein amounts in both cell lines (Fig-
JDP (J-domain-containing protein) (JDP-1 and JDP- ures 1b and 1d). Therefore 5 nM PMA was used for
2) subfamilies, which share structural similarities stimulation in all further experiments.
and form heterodimeric complexes with AP-1 pro-
teins (predominantly with Jun proteins), can bind − 727/ − 556 bp of the Axl promoter region is
to TRE (PMA-response element)-like sequences (5 - mandatory for PMA-induced promoter activity
TGAC/GTCA-3 ), as does AP-1. The net activity of To determine the minimal Axl promoter region
AP-1 can be regulated by changes in the transcrip- required for PMA stimulation, we transfected a
tion of genes encoding AP-1 subunits, by controlling series of deletion luciferase constructs into K562
the stability of their mRNAs, by post-translational cells. The AxlP1 construct ( − 1276/+7) exhib-
processing and turnover of pre-existing or newly syn- ited a maximum luciferase activity in both stim-
thesized AP-1 subunits and by specific interactions ulated and unstimulated conditions as compared
between AP-1 proteins and other transcription factors with pGL3-Basic (Figure 2a). Further deletion con-
and cofactors. The activation of AP-1 is involved in structs, AxlP2 ( − 1010/+7), AxlP3 ( − 727/+7),
cellular proliferation, apoptosis, differentiation and AxlP4 ( − 556/+7) and AxlP5 ( − 478/+7) showed

22 
C The Authors Journal compilation 
C 2011 Portland Press Limited
MAPK pathways in Axl gene expression Research article
Figure 1 PMA induces Axl gene expression
(a and c) K562 and HL-60 cells were stimulated with different concentrations of PMA for 24 h along with DMSO controls, and Axl
mRNA expression was determined by real-time PCR. GAPDH served as an internal control. (b and d) Both of the cell lines were
treated with 5 nM PMA or DMSO, or neither of them, as indicated. Cell lysates were prepared and Axl was detected by Western
blot analysis. Untreated or DMSO-treated cells served as controls and β-actin served as an internal loading control. TPA, PMA.

a gradual decrease in luciferase activity in both the 5 -flanking region located between − 727 and
unstimulated as well as PMA-stimulated condi- − 556 bp is required for PMA-stimulated Axl pro-
tions, and AxlP4 completely lost the ability to moter activity in leukaemia cells. Correspondingly,
be stimulated by PMA. AxlD1 ( − 2376/ − 556) a TRANSFAC database search and manual screen-
and AxlD2 ( − 2376/ − 770) lack the core promoter ing predicted AP-1/CREB [CRE (cAMP-response-
and, additionally, AxlD2 lacks a region spanning element)-binding protein]-binding motifs between
− 770/ − 556, this construct showing almost no luci- − 545 and − 530 bp, five overlapping TGCGTG
ferase activity, comparable with the pGL3-Basic vec- motifs between − 628 and − 606 bp, and a GT-
tor activity (Figure 2a) in both stimulated and un- rich sequence ( − 610 to − 589) in this region of
stimulated conditions. The − 2376/+7 reporter con- the Axl promoter (Figure 2b). Apart from these,
struct did not show any further significant induction one more AP-1 consensus sequence was predicted
in both conditions when compared with the AxlP1 at − 1030/ − 1024 bp of the Axl promoter.
( − 1276/+7) construct (results not shown), indicat-
ing that the main promoter activity for PMA stimula- AP-1 and 5 × TGCGTG repeats mediate
tion lies within the − 1276/+7 bp region, this being PMA-induced Axl promoter activity
comparable with the results seen for constitutive pro- To analyse the functional relevance of predicted
moter activity in Rko, HCT116 and HeLa cells (Mud- motifs in this important region of the pro-
duluru and Allgayer, 2008). From all of the deletion moter ( − 727/ − 556 bp), AP-1 putative motifs
experiments, we conclude that these data suggest that were mutated at four/five bases by site-directed

www.biolcell.org | Volume 103 (1) | Pages 21–33 23


G. Mudduluru and others

Figure 2 Delineating the 5 -flanking region essential for PMA-induced promoter activity
(a) Schematic representation of a series of deletion constructs of Axl, and reporter assays showing Axl promoter activity. In
total, 500 ng of Axl–luciferase constructs or pGL3-basic vector together with 25 ng of Renilla luciferase plasmid were transiently
transfected with Effectene into K562 cells. The firefly luciferase activity was normalized with Renilla luciferase activity. (b) Schem-
atic presentation of the Axl promoter, important for PMA stimulation with putative binding sites. (c) Schematic representation of
site-directed mutagenesis performed for AP-1 family-binding motifs in the Axl promoter region. Wild-type/mutant Axl promoter
construct (2 μg) along with the Renilla luciferase plasmid were transfected with the Nucleofector Kit V into K562 and HL-60 cell
lines. The wild-type unstimulated activity of AxlP1 was set to 100% (first bar), and the activity of other constructs was calculated
and plotted as a percentage of this value. The statistical difference of the activity of the mutated constructs in all panels is shown
against stimulated AxlP1 wild-type activity. *P  0.05. WT, wild-type. (d) AP-1 binding motifs in the Axl promoter.

mutagenesis within the AxlP1 ( − 1276 to +7) re- We observed that all mutants for single AP-1 mo-
porter plasmid. Different mutated constructs were tifs reduced the PMA-stimulated promoter activity
generated, with mutations either in single regions as compared with the wild-type AxlP1. Especially
(AP-1 I, AP-1 II, AP-1 III and AP-1 IV) or all the AP-1 II and AP-1 III mutant constructs signific-
of them (AP-1- I, II, III, IV) (Figures 2c and 2d). antly reduced the PMA-stimulated promoter activity
From our previous results, it was shown that the (P < 0.05), whereas AP-1 IV showed the trend (P =
Axl core promoter is driven by Sp1 and Sp3 in 0.058) in K562 cells. In HL-60 cells, the AP-1 III
colorectal cancer cell lines (Mudduluru and Allgayer, mutant construct showed significant reduction in the
2008). Therefore, to study the additional relevance promoter activity (P = 0.021), and others showed
of Sp family members under stimulated conditions, reduction in the promoter activity. The construct
we also used constructs mutated with Sp-binding do- where there was mutation of the four AP-1 motifs
mains of the Axl promoter (Sp1 abcde) for the present significantly reduced the PMA-stimulated promoter
study (Mudduluru and Allgayer, 2008). Mutated and activity in both of the cell lines (P = 0.0007 and P =
wild-type constructs were transfected into the K562 0.018 respectively). Unstimulated promoter activity
and HL-60 cells and compared for reporter activity. was not significantly altered with any of the AP-1

24 
C The Authors Journal compilation 
C 2011 Portland Press Limited
MAPK pathways in Axl gene expression Research article
mutations (Figure 2c). This confirms that all four for EMSA and supershift analysis to show the bind-
AP-1 motifs mediate PMA stimulation of the pro- ing of these transcription factors to the respective
moter, especially AP-1 II and III. However, they do cis-elements in the Axl promoter.
not affect basal promoter activity as compared with
the Sp family binding motifs (Sp1 abcde; Figure 2c).
In addition, the Sp1 abcde mutant construct did AP-1 family nuclear proteins bind
not show any significant increased promoter activ- to the Axl promoter
ity upon PMA stimulation (Figure 2c). EMSA (elec- To demonstrate that PMA-induced Axl gene ex-
trophoretic mobility-shift assay) and Western blot pression is dependent on AP-1 family transcription
analysis for Sp-binding motifs, and expression of factors binding to the promoter, gel-shift and super-
Sp1 and Sp3 after PMA stimulation did not show shift analysis was performed using nuclear extracts
an increase in binding efficiency and expression of either with DMSO- or PMA-stimulated K562 cells.
Sp1 and Sp3 (Supplementary Figures S1a and S1b EMSA and supershift analysis was performed with
at http://www.biolcell.org/boc/103/boc1030021add oligonucleotides corresponding to the AP-1-binding
.htm). This clearly implicates that the Sp-binding motifs (Figure 2d) and the GT-rich region. To de-
motifs are the essential mediators of basic promoter termine specific binding of AP-1 to these motifs, a
activity, whereas the AP-1 motifs are mediators of 50-fold excess of unlabelled wild-type, or a 50-fold
PMA-inducible promoter activity in leukaemia cells. excess of an oligonucleotide mutated for the respect-
ive AP-1-binding motif (Supplementary Figure S2
PMA increases the abundance at http://www.biolcell.org/boc/103/boc1030021add
of AP-1 family members .htm) were used for cold (non-radioactive) competi-
Functional and mutagenesis studies revealed that AP- tion. As shown in Supplementary Figure S2, the com-
1 motifs are important for PMA-stimulated Axl pro- petition with wild-type oligonucleotides completely
moter activity. To investigate whether this PMA- abolished specific binding of transcription factors,
induced promoter activity is due to an increase of ex- whereas, with the mutated oligonucleotides as com-
pression or stabilization of AP-1 transcription factors, petitors, the formation of specific complexes stayed
sequential Western blot analysis was performed for intact, demonstrating binding specificity for these
AP-1 and Axl proteins. Protein amounts of all AP-1 motifs.
members were increased in a time-dependent man- To investigate the binding of specific AP-1 fam-
ner, among them c-Fos being the one with an increase ily members to these motifs, a comparative super-
after 1 h of stimulation. Starting at 2 h, Fos B pro- shift analysis with all AP-1 family members was per-
tein increased, with a maximum level at 4 h. After formed with nuclear extracts of K562 cells, with or
30 min of stimulation, activated c-Jun was observed, without PMA stimulation (Figure 2d). PMA stim-
and this was constantly present for 24 h. In addition, ulation increased the abundance of specific com-
total protein levels of c-Jun were increased, starting plexes to the AP-1 motifs. Incubation of AP-1
at 2 h. Axl protein levels were increased after 4 h of family antibodies identified phospho-c-Jun within
treatment and were further increased at 24 h (Fig- the AP-1 complexes at all four AP-1 motifs and
ure 3a). Most of the AP-1 family members’ gene ex- at the GT-rich region, under PMA-unstimulated
pression was induced by PMA-stimulation in K562 conditions (Figures 3b and 3c, and Supplement-
cells. Activated c-Jun levels were stable from 8 to ary Figures S3a and S3b, top panel at http://www.
24 h after stimulation, which was paralled by an in- biolcell.org/boc/103/boc1030021add.htm). Follow-
crease of Axl protein levels at 8 h and a further 4-fold ing PMA stimulation, Fos B and Fra 1 were iden-
increase of Axl protein at 24 h. Taken together, these tified to be increased within the AP-1 complexes at
data show that PMA activates c-Jun and stabilizes all sites, and also at the GT-rich region (Figures 3b
the expression of all AP-1 family members, which and 3c, and Supplementary Figures S3a and S3b,
is followed by an increase of Axl protein expression bottom panel). Additionally, Jun B was identified
in K562 cells. Since we observed a higher activa- within complexes bound to the AP-1 I and AP-1 IV
tion/expression of AP-1 transcription factors at 8 h motif, and Jun B, Jun D and c-Fos within complexes
post-stimulation with PMA, we used this time point at the AP-1 II and AP-1 III motif (Figures 3b and

www.biolcell.org | Volume 103 (1) | Pages 21–33 25


G. Mudduluru and others

Figure 3 AP-1 family members bind to the Axl promoter, and their abundance increases after PMA stimulation
(a and b) Supershift analysis was performed with (bottom panel) or without (top panel) PMA stimulation (for 8 h) and nuclear
extracts of K562, for AP-1 family members (c-Jun, Jun B, Jun D, c-Fos, Fos B, Fra 1 and Fra 2) using AP-1 I ( − 628/ − 606 bp)
and AP-1 II and III ( − 655/ − 640 bp) as radioactive probes. IgG served as the negative control. Specific supershifted complexes
are indicated with arrows. (c) K562 cells were stimulated with 5 nM PMA for the time points indicated, followed by preparation
of total cell lysates. Untreated cells were used as a control. Phospho-c-Jun, c-Jun, Jun B, Jun D, c-Fos, Fos B, Fra 1, Fra 2
and Axl protein levels were determined by Western blot analysis. β-Actin served as the internal control. The results shown are
representative of two similar experiments. (d) The in vivo association of phospho-c-Jun with the Axl promoter was evaluated with
a ChIP assay in K562 and HL-60 cells after 24 h of DMSO, PMA, Gö6983, or Gö6983 with PMA treatment. Immunoprecipitated
DNA with the specific antibody was amplified by real-time PCR (SYBR® Green) using specific primers. TPA, PMA.

3c, and Supplementary Figures S3a and S3b, bottom and PMA-stimulated supershift bands for CREB and
panel). phospho-CREB; however, an intense shifted band was
The AP-1 II and AP-1 III motifs harbour CRE and observed with phospho-c-Jun following PMA stim-
TRE consensus sequences, which could bind CREB ulation (Supplementary Figure S4b at http://www.
and AP-1 transcription factors respectively. To define biolcell.org/boc/103/boc1030021add.htm), even
binding of phospho-CREB and phospho-c-Jun tran- though increased activated p-CREB levels were
scription factors together at these motifs, supershift observed within nuclear proteins (Supplementary
analysis was performed. No significant differences in Figure S4a). These results suggest that AP-1 family
band intensities were observed between unstimulated transcription factors are binding preferentially to the

26 
C The Authors Journal compilation 
C 2011 Portland Press Limited
MAPK pathways in Axl gene expression Research article
specific AP-1 motifs in the Axl promoter, as opposed between untreated and DMSO-treated samples.
to CREB transcription factors. PMA-induced Axl gene expression was completely
ChIP (chromatin immunoprecipitation) analysis abolished by the PKC inhibitor Gö6983 in paral-
was performed to determine the in vivo relevance of lel with the downstream signalling molecules ERK,
AP-1 transcription factor binding to the Axl pro- p38, JNK and phospho-c-Jun, in both of the cell
moter, comparing DMSO treatment with PMA stim- lines (Figures 4a and 4b). Specific inhibitors of ERK
ulation, and, to get initial insights into putative (U0216), p38 (SB202190) and JNK (SP600125) par-
signalling mediators, with a PMA/PKC inhibitor tially abolished PMA-induced Axl gene expression
in K562 and HL-60 cells. A non-specific region of in K562 cells (Figure 4a). Both the ERK1/2 and
the uPAR [uPA (urokinase-type plasminogen activ- p38 inhibitors completely abolished Axl gene ex-
ator) receptor] promoter was used as a negative con- pression in the HL-60 cell line (Figure 4a). Further-
trol (Mudduluru and Allgayer, 2008), which did not more, Western blot analysis showed similar results in
show any specific changes after treatment (results K562 cells (Figures 4c and 4d). Individual inhibitors
not shown). PMA stimulation increased the bind- of JNK/ERK/p38 could not abolish the activation of
ing of phospho-c-Jun to the Axl promoter; also, 1 h c-Jun completely, as indicated by Western blotting
pre-treatment with Gö6983 decreased the binding for phosphorylated c-Jun (Figures 4c and 4d). This
of phospho-c-Jun (Figure 3d). Similar results were could be due to the leakage of signalling through
observed in Western blot analysis; Gö6983 inhibited other MAPK signalling molecules. This suggests that
c-Jun activation (Figure 4b). These data support the PMA-induced signalling might be working mainly
notion that, in vivo, AP-1 binds to the predicted AP- through ERK and p38. However, the differences in
1 consensus and GT-rich region of the Axl promoter PMA-induced Axl gene expression through MAPK
upon PMA-stimulation, and that the PKC pathway signalling in these cell lines need to be further invest-
might be one important mediator. igated thoroughly. Apart from the findings presented,
post-transcriptional regulation of Axl cannot be ruled
The PKC/MAPK signalling pathway mediates out after PMA stimulation. Furthermore, to gain ad-
PMA-induced Axl gene expression ditional information on the PMA-induced effect on
To investigate signalling molecules mediating PMA- basal Axl gene expression, mithramycin, which has
induced Axl gene expression, luciferase assay, EMSA, already been shown to abolish Axl basal gene expres-
quantitative PCR and Western blot analysis were per- sion via the prevention of Sp binding in colorectal
formed following treatment of the cells with various cell lines (Mudduluru and Allgayer, 2008), was in-
chemical inhibitors of the MAPK signalling pathway cluded. PMA-activated MAPK could not induce Axl
prior to PMA stimulation. K562 and HL-60 cells gene expression after mithramycin treatment (Fig-
were transfected with AxlP1 reporter constructs and ure 4c), even though the MAPK signalling cascade
treated with the specific inhibitors for 45 min and sti- was active, suggesting that basal Axl gene expres-
mulated with PMA for 24 h (Figure 5a). PKC and sion via Sp factors is a prerequisite for inducible ex-
ERK1/2 inhibitors significantly reduced the Axl pro- pression. Taken together, these results suggest that
moter activity, whereas a p38 inhibitor showed the PMA-induced Axl gene expression is mainly regu-
trend and a JNK inhibitor showed reduced promoter lated via PKC and MAPK/ERK1/2 and MAPK/p38
activity. Similar results were observed with EMSA pathways, and it further reconfirms in leukaemia cells
analysis with inhibitor treatment (Figure 5b). PKC, that Sp family transcription factors are required for
ERK1/2 and p38 inhibitors showed reduced PMA- constitutive Axl gene expression.
induced AP-1 binding to the Axl promoter. Addi-
tionally, K562 or HL-60 cells were untreated, treated
with DMSO or treated with PMA following 45 min Discussion
of pretreatment with PKC/JNK/ERK/p38 inhibit- Axl has been discussed as a transforming gene in
ors, or the GC-rich binding compound mithramy- leukaemia (Janssen et al., 1991; O’Bryan et al.,
cin (Figure 4). No differences in the activation and 1991); however, mechanisms inducing its expres-
expression of PKC signalling molecules and in sion upon oncogenic stimuli in leukaemia are still
Axl expression and protein amounts were observed the subject of investigation. In the present study

www.biolcell.org | Volume 103 (1) | Pages 21–33 27


G. Mudduluru and others

Figure 4 The MAPK pathway via different signalling molecules mediates PMA-induced Axl gene expression
(a) Real-time PCR analysis of K562 and HL-60 cells were performed with DMSO, PMA or with specific inhibitors in combination
with PMA, as indicated. Cells were pretreated with inhibitors for 45 min and further stimulated with 5 nM PMA for 24 h. (b) Western
blot analysis of K562 and HL-60 cells were performed with DMSO, PMA, Gö6983 or a combination of PMA and Gö6983. Cells
were pretreated with DMSO or Gö6983 in the respective wells for 45 min and stimulated for either 1 h or 24 h along with PMA.
Specific antibodies against the indicated molecules were used, together with anti-β-actin as a loading control. (c and d) Effect
of inhibitors on PMA-induced Axl protein expression. K562 cells were pretreated with ERK1/2 inhibitor U0216 (10 μM), p38
inhibitor SB202190 (10 μM), JNK inhibitor SP600125 (5 to 10 μM) and mithramycin (100 nM) 45 min before stimulation with
5 nM PMA and incubated for an additional 24 h along with inhibitors or as indicated. Untreated and DMSO-treated cells served
as experimental controls. Cell lysates were prepared, and phosphorylated (phospho-ERKs, phospho-p38, phospho-JNK and
phospho-c-Jun) and total (ERKs, p38, JNK, Axl) proteins were detected by Western blot analysis. β-Actin served as the internal
control. TPA, PMA.

we show that, in leukaemia cells, PMA-induced Axl ily members and induces the expression/stabilization
gene expression is mediated by AP-1 family tran- of AP-1 family transcription factors that lead to
scription factors bound to AP-1 cis-acting elements induced Axl gene expression in leukaemia cell
within − 660 and − 580 bp of the promoter region. lines. Moreover, we found that PMA-stimulated Axl
This region is essential for PMA-induced Axl pro- gene expression is mediated through ERK/JNK/p38
moter activity. PMA-stimulation activates AP-1 fam- signalling.

28 
C The Authors Journal compilation 
C 2011 Portland Press Limited
MAPK pathways in Axl gene expression Research article
Figure 5 The MAPK pathway inhibitors reduce Axl promoter activity and AP-1 binding to its promoter
(a) Luciferase analysis of AxlP1 either with or without pre-treatment of specific inhibitors in K562 and HL-60 cells. (b) EMSA
analysis was performed with AP-1 II and III ( − 655/ − 640 bp) motif with or without PMA stimulation (8 h) after 45 min of
pre-treatment with the specific inhibitors in K562 and HL-60 cells. TPA, PMA.

AP-1 consists of various dimers of the Fos (c-Fos, tivation itself can be mediated by AP-1 binding to
FosB, Fra-1 and Fra-2), Jun (c-Jun, JunB and JunD), TRE elements within the promoter, for example of
and CREB/ATF protein families, as well as other bZip c-Jun and Fra-1, inducing their expression (Angel
proteins. In addition, associations have been observed et al., 1988; Bergers et al., 1995). The − 1276/+7
between Fos or Jun and the p65 subunit of NF-κB region of the Axl promoter showed maximum activ-
(nuclear factor κB) (Stein et al., 1993) and ATF-2 ity under PMA-stimulated conditions (Mudduluru
and p50-NF-κB (Du et al., 1993). AP-1 transcrip- and Allgayer, 2008). A TRANSFAC database search,
tion factor heterodimers or homodimers bind to a 5 delineation reporter analysis, site-directed muta-
consensus cis-element in the promoters of a number genesis, EMSA and supershift analysis of the Axl pro-
of genes (Adler et al., 1994). Evidence for the critical moter identified two unique binding motifs of AP-1-
importance of AP-1 activity in malignant transform- and TRE-consensus sequences, which are essential
ation induced by tumour promoters or by oncogenes for PMA-stimulated promoter activity (AP-1 II and
has been reported (Bernstein and Colburn, 1989; AP-1 III). Apart from these two, AP-1-binding mo-
Domann et al., 1994; Dong et al., 1994). The regula- tifs containing partially overlapping five TGCGTG
tion of AP-1 activity is complex and occurs at differ- repeats, a − 607 to − 589 bp unique GT-rich
ent levels, including dimer composition, transcrip- motif and a distant − 1010 to − 1022 bp AP-1 mo-
tional and post-translational events, and interaction tif show specific binding complexes of AP-1 tran-
with ancillary proteins. AP-1 is induced by several scription factors under PMA-stimulated conditions,
external stimuli that increase MAPK activity, indu- which are also required for stimulated Axl promoter
cing activation of c-Fos and c-Jun. PMA, a tumour- activity. Axl protein amounts could be detected at 4 h
promoting agent, often serves as a model agent to and increased gradually up to 24 h, whereas activated
study the mechanism by which growth factors reg- c-Jun was detected at 30 min and achieved maximum
ulate growth and differentiation of the cells, and is activated levels at 12 h. Among AP-1 members, an
an endogenous activator of PKC and MAPK path- induction of c-Fos and c-Jun was detected as early
ways (Jaken, 1996; Parekh et al., 2000). AP-1 ac- as 1 h and 2 h respectively, and achieved maximum

www.biolcell.org | Volume 103 (1) | Pages 21–33 29


G. Mudduluru and others

levels 4–12 h after PMA treatment. These findings activated MAPK, activated c-Jun and promoted Axl
suggest that c-Jun is being activated at early time promoter activity and gene expression (Figure 4),
points upon PMA treatment and, indeed, transactiv- indicating that activation of the MAPK cascade is
ates/stabilizes the other AP-1 transcription factors. relevant for PMA-induced Axl gene expression (An-
Thus c-Jun or c-Fos might be playing a major role in gel and Karin, 1991; Minden and Karin, 1997; Kast
the earlier course and the rest of the AP-1 factors may et al., 2003). Diverse inhibitors of different molecules
play a major role in the later course of the induction of MAPK-associated pathways [JNK (SP600125),
of Axl gene transcription by PMA, a phenomenon ERK1/2 (U0126), and p38 (SB202190)] repressed
which has been described previously, e.g. for Fra1 c-Jun activation and reduced AP-1-binding com-
and proenkephalin (Adiseshaiah et al., 2003; Won plexes to the Axl promoter, indicating three dif-
et al., 1998). We can speculate that upon PMA stim- ferent lines of signalling in this particular situation
ulation, AP-1 transcription factors are activated and (Pulverer et al., 1991; Papavassiliou et al., 1995; Shin
autoregulated or stabilized at their protein levels, and et al., 2002; Kast et al., 2003). These findings sug-
then transactivate Axl gene expression. gest that inhibition of PKC or downstream signalling
c-Jun transcriptionally regulates EGFR [EGF molecules of PKC (ERK1/2, JNK, p38) activity res-
(epidermal growth factor) receptor] and HB-EGF ults in an inhibition of AP-1 transactivation, which is
(heparin-binding EGF-like growth factor), thereby an important mechanism for Axl transactivation and
controlling keratinocyte proliferation and skin tu- gene expression under tumorigenic PMA stimula-
mour formation (Zenz et al., 2003). Hu et al. tion, suggesting therapeutic considerations for leuk-
(1994) and Kustikova et al. (1998) reported that aemia. Mithramycin treatment completely blocked
AP-1 transcription factors stimulate the transcrip- PMA-induced Axl gene expression, even though the
tion of MMP (matrix metalloproteinase) 9, MMP1, MAPK signalling cascade was active, which is a con-
uPA, uPAR, HMGI(Y) [high-mobility group I (Y)] firmation that, also in leukaemia, Sp1/Sp3 is essential
and E-cadherin, which are tumour progression- and for the basal transcriptional activation of Axl gene
metastasis-associated genes. Since Axl overexpression expression, which is a prerequisite for inducibility
is associated with increased invasion, metastasis and (Mudduluru and Allgayer, 2008).
angiogenesis in various cancer cells, and also its over- In summary, we have identified an AP-1 family
expression is reported in different cancers, such as binding region − 660 to − 580 bp within the Axl
metastatic colon and prostate carcinoma, gastric, en- promoter that is indispensable for PMA-induced
dometrial, certain types of breast cancers and sarco- Axl gene expression in leukaemia cells. Induction of
mas (Neubauer et al., 1994; Weiner et al., 1994; transcription factor AP-1 binding is mainly mediated
Craven et al., 1995; Jacob et al., 1999; Meric et al., through the ERK1/2/p38 pathway, and phorbol es-
2002; Wu et al., 2002; Sun et al., 2003; Budagian ters transactivate and/or stabilize the expression of all
et al., 2005b; Shieh et al., 2005; Vajkoczy et al., AP-1 family transcription factors in leukaemia cells.
2006), the transactivation of Axl by AP-1 following
PMA stimulation adds to the list of essential AP-1
target genes in the context of carcinogenesis, inva- Materials and methods
Cell culture, reagents and treatments
sion, metastasis and angiogenesis.
K562 and HL-60 cells were obtained from the A.T.C.C. and
Signals transmitted through PKC can potentially grown at 37◦ C in RPMI medium supplemented with 10%
activate the network of MAPK signal-transduction FCS (fetal calf serum). Mock-treated control cells were handled
pathways (ERK1/2, JNK, p38, ERK5 and ERK3/4), identically with PMA (Sigma)-stimulated cells with the excep-
which play an important role in regulating the re- tion that only medium was added. For inhibition studies, cells
were treated with or without the following inhibitors for 45
sponse of cells to various extracellular stimuli, such min: 5 μM Gö6983 (Calbiochem), 10 μM U0216 (Calbiochem),
as PMA (Kast et al., 2003; Rubinfeld and Seger, 10 μM SB202190 (Calbiochem), 100 nM mithramycin (Sigma)
2004; Kolch et al., 2005). The conventional posi- and 5 or 10 μM SP600125 (Calbiochem) respectively.
tion of c-Jun/AP-1 in signal transduction cascades
Preparation of protein extracts and immunoblotting
has been downstream of MAPK pathways. Thus we Protein lysates were prepared as described previously by Mud-
studied the effect of inhibitors of each MAPK path- duluru et al. (2008). Briefly, for immunoblotting, samples
way on PMA-dependent Axl gene expression. PMA- (50 μg/lane) were boiled for 5 min, separated via SDS/PAGE,

30 
C The Authors Journal compilation 
C 2011 Portland Press Limited
MAPK pathways in Axl gene expression Research article
and transferred on to PVDF membranes. After transfer, the mem- acrylamide gel in 0.25 × Tris/borate/EDTA buffer at 10 V/cm
branes were blocked with 5% non-fat skimmed milk containing for 3.5 h. The gel was dried and analysed by autoradiography.
TTBS [TBS (Tris-buffered saline, 50 mM Tris/HCl and 150 mM
NaCl, pH 8.0) with 0.1% Tween] for 3 h at room temperature
(21◦ C) and then probed with primary antibodies against c-Jun Generation of luciferase reporter constructs
(Santa Cruz Biotechnology, sc-45x), phospho-c-Jun (Santa Cruz Luciferase reporter constructs of the Axl promoter gener-
Biotechnology, sc-822), Jun B (Santa Cruz Biotechnology, sc- ated as described by Mudduluru et al. (2008). Two addi-
46x), Jun D (Santa Cruz Biotechnology, sc-74x), c-Fos (Santa tional reporter constructs were generated (Mudduluru and
Cruz Biotechnology, sc-52x), Fos B (Santa Cruz Biotechno- Allgayer, 2008), lacking the transcriptional start site
logy, sc-7203x), Fra1 (Santa Cruz Biotechnology, sc-22794x), and the core promoter, using specific reverse primers. The
Fra2 (Santa Cruz Biotechnology, sc-604x), phospho-Jnk (Cell primer sequences are listed in Supplementary Table S2 (at
Signaling Technology, cs-9251s), JNK (Cell Signaling Techno- http://www.biolcell.org/boc/103/boc1030021add.htm). Mutant
logy, cs 9252), phospho-ERK1/2 (Cell Signaling Technology, cs constructs were generated using the QuikChange® XL site-
9101s), ERK1/2 (Cell Signaling Technology, cs 9102), phospho- directed mutagenesis kit from Stratagene using AxlP1 ( − 1276
p38 (Cell Signaling Technology, cs 9211s), p38 (Cell Signaling to +7) as a template. The sequences of the mutant oligonuc-
Technology, cs-9212), Axl (Santa Cruz Biotechnology, sc-1096) leotides are shown in Supplementary Table S1. Successful incor-
or β-actin (Santa Cruz Biotechnology, sc-1616) for 2 h at room poration of the mutations was confirmed by automated sequen-
temperature. After three washes with TTBS, blots were incub- cing.
ated with the appropriate secondary antibodies conjugated with
horseradish peroxidase. After final washes with TTBS, the mem- Transfection and luciferase assays
branes were exposed to film after ECL (enhanced chemilumines- For luciferase assays, 0.5 × 106 K562 cells were plated in 24-well
cence; Amersham Biosciences). plates at the time of transfection. Luciferase reporter plasmids
(0.5 μg of each) were transfected with the Effectene® transfection
reagent (Qiagen). For AP-1/Sp family binding motif mutation
RNA isolation and RT (reverse transcriptase)–PCR and specific inhibitor-treated luciferase assay analysis, cells were
Total RNA was extracted using the RNeasy Midi kit electroporated with the Amaxa Cell Line Nucleofector Kit V
(Qiagen) according to the manufacturer’s instructions. Total (Lonza) according to the manufacturer’s protocol into 2 × 106
RNA (1 μg) was reverse-transcribed by random hexamer cells with 2 μg of respective reporter plasmid. For all luciferase
primers using SuperScript II reverse transcriptase (Invitro- reporter assays, 25 ng of pRL-TK (Renilla luciferase-thymidine
gen). The single-strand cDNA was amplified by SYBR® Green kinase; Promega) was also co-transfected and measured to nor-
quantitative PCR using specific primers as described previ- malize transfection efficiency. After 12 h of transfection, cells
ously (Mudduluru et al., 2010). GAPDH (glyceraldehyde-3- were either pre-treated with specific inhibitors for 45 min and/or
phosphate dehydrogenase) was used as an internal control. stimulated with 5 nM of PMA. After 24 h, cells were washed
The sequences of the sense and antisense primers used for twice with PBS and lysed with 100 μl of passive lysis buffer
either semi-quantitative, or SYBR® Green quantitative PCR (Promega) for 20 min. Luciferase activity of 20 μl of cell lysate
were: GAPDH (sense, 5 -GTCTTCACCACCATGGAGAA-3 was measured via the dual-luciferase reporter assay system (Pro-
and antisense, 5 -ATCCACAGTCTTCTGGGTGG-3 ) and Axl mega) according to the manufacturer’s instructions. Assays for
(sense, 5 -GGTGGCTGTGAAGACGATGA-3 and antisense, all samples were performed in triplicate, and the results were
5 -CTCAGATACTCCATGCCACT-3 ). averaged.

ChIP assay
Preparation of nuclear extract and EMSA ChIP assays were performed as described previously (Leupold
Cells were either pre-treated with the respective specific et al., 2007; Mudduluru and Allgayer, 2008) using either
inhibitor for 45 min and/or treated for 8 h with PMA anti-phospho-c-Jun (Santa Cruz Biotechnology, sc-822 X) and
(5 nM), then cells were harvested into 50 ml Falcon tubes anti-IgG (as a control; Santa Cruz Biotechnology, sc-2338 X)
and washed with ice-cold PBS. Nuclear extraction, protein antibodies. K562 or HL-60 cells were either mock-treated with
concentration estimation and EMSAs were essentially per- DMSO or treated with Gö6983 (5 μM) in the respective samples
formed as described previously (Mudduluru and Allgayer, 45 min prior to PMA (5 nM) stimulation. After 24 h, cells were
2008; Mudduluru et al., 2010). Briefly, 100 ng of annealed processed as described previously (Leupold et al., 2007; Muddu-
double-stranded oligonucleotides, GT-rich ( − 609/ − 569), luru and Allgayer, 2008). Phospho-c-Jun-binding intensity to
AP-1 I (5 × TGCGTG repeats: − 635/ − 601), AP-1 II the Axl promoter was measured using SYBR® Green PCR with
and III ( − 672/ − 631), and AP-1 IV ( − 1036/ − 1008), specific primers: sense 5 -AGTGTGTCTGTGGGCCAGTA-3
of the Axl promoter listed in Supplementary Table S1 and antisense 5 -CCTTCATTCCCCCTCACTCT-3 .
(at http://www.biolcell.org/boc/103/boc1030021add.htm) were
end-labelled with [γ -33 P]dATP using T4 polynucleotide kinase
(NEB), and purified with Sephadex G-50 columns (Amersham Statistical analysis
Bioscience). The DNA-binding reaction was conducted on ice for Statistical analyses were performed using SPSS statistical soft-
20 min with 5 μg of K562 cell nuclear extract. For supershift as- ware. A Student’s t test was performed for data with normal
says, antibodies were incubated with the reaction mixture at 4◦ C distribution, and the Wilcoxon test for all other analyses. P
for an additional 60 min. Samples were analysed on a 5% poly- values of 0.05 were considered as statistically significant.

www.biolcell.org | Volume 103 (1) | Pages 21–33 31


G. Mudduluru and others

Author contribution Budagian, V., Bulanova, E., Orinska, Z., Duitman, E., Brandt, K.,
Concept and design of the research was by Girid- Ludwig, A., Hartmann, D., Lemke, G., Saftig, P. and Bulfone-Paus,
S. (2005a) Soluble Axl is generated by ADAM10-dependent
har Mudduluru. The experiments were carried out cleavage and associates with Gas6 in mouse serum. Mol. Cell.
by Giridhar Mudduluru and Jörg Leupold; collec- Biol. 25, 9324–9339
tion and/or assembly of data was by Giridhar Mud- Budagian, V., Bulanova, E., Orinska, Z., Thon, L., Mamat, U.,
duluru; and data analysis and interpretation was by Bellosta, P., Basilico, C., Adam, D., Paus, R. and Bulfone-Paus, S.
Bulfone-Paus (2005b) A promiscuous liaison between IL-15
Giridhar Mudduluru and Philipp Stroebel. The ma- receptor and Axl receptor tyrosine kinase in cell death control.
nuscript was written by Giridhar Mudduluru and EMBO J. 24, 4260–4270
Heike Allgayer. Financial support for the study was Burchert, A., Attar, E.C., McCloskey, P., Fridell, Y.W. and Liu, E.T.
obtained by Heike Allgayer. All authors approved the (1998) Determinants for transformation induced by the Axl receptor
tyrosine kinase. Oncogene 16, 3177–3187
final manuscript. Costa, M., Bellosta, P. and Basilico, C. (1996) Cleavage and release
of a soluble form of the receptor tyrosine kinase ARK in vitro and in
vivo. J. Cell Physiol. 168, 737–744
Acknowledgements Craven, R.J., Xu, L.H., Weiner, T.M., Fridell, Y.W., Dent, G.A.,
This publication contains parts of the dissertation Srivastava, S., Varnum, B., Liu, E.T. and Cance, W.G. (1995)
(Dr. sc. hum.) of Giridhar Mudduluru at the Man- Receptor tyrosine kinases expressed in metastatic colon cancer.
Int. J. Cancer 60, 791–797
nheim Faculty of Medicine, University of Heidelberg, Demarchi, F., Verardo, R., Varnum, B., Brancolini, C. and Schneider,
Mannheim, Germany. We thank Erika Hillerich and C. (2001) Gas6 anti-apoptotic signaling requires NF-κB activation.
Laura Nelson for excellent help and critical appraisal J. Biol. Chem. 276, 31738–31744
of the manuscript prior to submission. Domann, F.E., Levy, J.P., Birrer, M.J. and Bowden, G.T. (1994) Stable
expression of a c-JUN deletion mutant in two malignant mouse
epidermal cell lines blocks tumor formation in nude mice. Cell
Growth Differ. 5, 9–16
Funding Dong, Z., Birrer, M.J., Watts, R.G., Matrisian, L.M. and Colburn, N.H.
H.A. was supported by the Alfried Krupp von (1994) Blocking of tumor promoter-induced AP-1 activity inhibits
Bohlen und Halbach Foundation (Award for Young induced transformation in JB6 mouse epidermal cells. Proc. Natl.
Full Professors), Essen; the Hella-Bühler-Foundation, Acad. Sci. U.S.A. 91, 609–613
Du, W., Thanos, D. and Maniatis, T. (1993) Mechanisms of
Heidelberg; the Dr Ingrid zu Solms Foundation, transcriptional synergism between distinct virus-inducible
Frankfurt/Main; the Hector Foundation, Weinheim, enhancer elements. Cell 74, 887–898
Germany; the FRONTIER Excellence Initiative of Eferl, R. and Wagner, E.F. (2003) AP-1: a double-edged sword in
the University of Heidelberg; and the Walter Schulz tumorigenesis. Nat. Rev. Cancer 3, 859–868
Graham, D.K., Dawson, T.L., Mullaney, D.L., Snodgrass, H.R. and
Foundation, Munich, Germany. Earp, H.S. (1994) Cloning and mRNA expression analysis of a
novel human protooncogene, c-mer. Cell Growth Differ. 5, 647–657
Hu, E., Mueller, E., Oliviero, S., Papaioannou, V.E., Johnson, R. and
References Spiegelman, B.M. (1994) Targeted disruption of the c-fos gene
Adiseshaiah, P., Papaiahgari, S.R., Vuong, H., Kalvakolanu, D.V. and demonstrates c-fos-dependent and -independent pathways for
Reddy, S.P. (2003) Multiple cis-elements mediate the gene expression stimulated by growth factors or oncogenes.
transcriptional activation of human fra-1 by EMBO J. 13, 3094–3103
12-O-tetradecanoylphorbol-13-acetate in bronchial epithelial cells. Jacob, A.N., Kalapurakal, J., Davidson, W.R., Kandpal, G., Dunson,
J. Biol. Chem. 278, 47423–47433 N., Prashar, Y. and Kandpal, R.P. (1999) A receptor tyrosine kinase,
Adler, V., Unlap, T. and Kraft, A.S. (1994) A peptide encoding the UFO/Axl, and other genes isolated by a modified differential
c-Jun delta domain inhibits the activity of a c-jun amino-terminal display PCR are overexpressed in metastatic prostatic carcinoma
protein kinase. J. Biol. Chem. 269, 11186–11191 cell line DU145. Cancer Detect Prev. 23, 325–332
Angel, P., Hattori, K., Smeal, T. and Karin, M. (1988) The jun
Jaken, S. (1996) Protein kinase C isozymes and substrates. Curr.
proto-oncogene is positively autoregulated by its product,
Opin. Cell Biol. 8, 168–173
Jun/AP-1. Cell 55, 875–885
Janssen, J.W., Schulz, A.S., Steenvoorden, A.C., Schmidberger, M.,
Angel, P. and Karin, M. (1991) The role of Jun, Fos and the AP-1
Strehl, S., Ambros, P.F. and Bartram, C.R. (1991) A novel putative
complex in cell-proliferation and transformation. Biochim. Biophys.
Acta 1072, 129–157 tyrosine kinase receptor with oncogenic potential. Oncogene 6,
Bergers, G., Graninger, P., Braselmann, S., Wrighton, C. and 2113–2120
Busslinger, M. (1995) Transcriptional activation of the fra-1 gene by Kast, C., Wang, M. and Whiteway, M. (2003) The ERK/MAPK
AP-1 is mediated by regulatory sequences in the first intron. Mol. pathway regulates the activity of the human tissue factor pathway
Cell. Biol. 15, 3748–3758 inhibitor-2 promoter. J. Biol. Chem. 278, 6787–6794
Bernstein, L.R. and Colburn, N.H. (1989) AP1/jun function is Kolch, W., Calder, M. and Gilbert, D. (2005) When kinases meet
differentially induced in promotion-sensitive and resistant JB6 mathematics: the systems biology of MAPK signalling. FEBS Lett.
cells. Science 244, 566–569 579, 1891–1895

32 
C The Authors Journal compilation 
C 2011 Portland Press Limited
MAPK pathways in Axl gene expression Research article
Kustikova, O., Kramerov, D., Grigorian, M., Berezin, V., Bock, E., Rubinfeld, H. and Seger, R. (2004) The ERK cascade as a prototype
Lukanidin, E. and Tulchinsky, E. (1998) Fra-1 induces of MAPK signaling pathways. Methods Mol. Biol. 250, 1–28
morphological transformation and increases in vitro invasiveness Shaulian, E. and Karin, M. (2002) AP-1 as a regulator of cell life and
and motility of epithelioid adenocarcinoma cells. Mol. Cell. Biol. 18, death. Nat. Cell Biol. 4, E131–E136
7095–7105 Shieh, Y.S., Lai, C.Y., Kao, Y.R., Shiah, S.G., Chu, Y.W., Lee, H.S. and
Lai, C., Gore, M. and Lemke, G. (1994) Structure, expression, and Wu, C.W. (2005) Expression of axl in lung adenocarcinoma and
activity of Tyro 3, a neural adhesion-related receptor tyrosine correlation with tumor progression. Neoplasia 7, 1058–1064
kinase. Oncogene 9, 2567–2578 Shin, M., Yan, C. and Boyd, D. (2002) An inhibitor of c-jun amino
Leupold, J.H., Asangani, I., Maurer, G.D., Lengyel, E., Post, S. and terminal kinase (SP600125) represses c-Jun activation,
Allgayer, H. (2007) Src induces urokinase receptor gene expression DNA-binding and PMA-inducible 92-kDa type IV collagenase
and invasion/intravasation via activator protein-1/p-c-Jun in expression. Biochim. Biophys. Acta 1589, 311–316
colorectal cancer. Mol. Cancer Res. 5, 485–496 Stein, B., Baldwin, Jr, A.S., Ballard, D.W., Greene, W.C., Angel, P. and
Meric, F., Lee, W.P., Sahin, A., Zhang, H., Kung, H.J. and Hung, M.C. Herrlich, P. (1993) Cross-coupling of the NF-κB p65 and Fos/Jun
(2002) Expression profile of tyrosine kinases in breast cancer. Clin. transcription factors produces potentiated biological function.
Cancer Res. 8, 361–367 EMBO J. 12, 3879–3891
Minden, A. and Karin, M. (1997) Regulation and function of the JNK Sun, W.S., Fujimoto, J. and Tamaya, T. (2003) Coexpression of
subgroup of MAP kinases. Biochim. Biophys. Acta 1333, F85–F104 growth arrest-specific gene 6 and receptor tyrosine kinases Axl
Mudduluru, G. and Allgayer, H. (2008) The human receptor tyrosine and Sky in human uterine endometrial cancers. Ann. Oncol. 14,
kinase Axl gene–promoter characterization and regulation of 898–906
constitutive expression by Sp1, Sp3 and CpG methylation. Biosci. Vajkoczy, P., Knyazev, P., Kunkel, A., Capelle, H.H., Behrndt, S., von
Rep. 28, 161–176 Tengg-Kobligk, H., Kiessling, F., Eichelsbacher, U., Essig, M.,
Mudduluru, G., Vajkoczy, P. and Allgayer, H. (2010) Myeloid zinc Read, T.A. et al. (2006) Dominant-negative inhibition of the Axl
finger 1 induces migration, invasion, and in vivo metastasis through receptor tyrosine kinase suppresses brain tumor cell growth and
Axl gene expression in solid cancer. Mol. Cancer Res. 8, 159–169 invasion and prolongs survival. Proc. Natl. Acad. Sci. U.S.A. 103,
Neubauer, A., Fiebeler, A., Graham, D.K., O’Bryan, J.P., 5799–5804
Schmidt, C.A., Barckow, P., Serke, S., Siegert, W., Sndograss, Weiner, T.M., Liu, E.T., Craven, R.J. and Cance, W.G. (1994)
H.R., Huhn, D. et al. (1994) Expression of axl, a transforming Expression of growth factor receptors, the focal adhesion kinase,
receptor tyrosine kinase, in normal and malignant hematopoiesis. and other tyrosine kinases in human soft tissue tumors. Ann. Surg.
Blood 84, 1931–1941 Oncol. 1, 18–27
O’Bryan, J.P., Frye, R.A., Cogswell, P.C., Neubauer, A., Kitch, B., Won, J.S., Song, D.K., Kim, Y.H., Huh, S.O. and Suh, H.W. (1998) The
Prokop, C., Espinosa, 3rd, R., Le Beau, M.M., Earp, H.S. and stimulation of rat astrocytes with phorbol-12-myristate-13-acetate
Liu, E.T. (1991) Axl, a transforming gene isolated from primary increases the proenkephalin mRNA: involvement of
human myeloid leukemia cells, encodes a novel receptor tyrosine proto-oncogenes. Brain Res. Mol. Brain Res. 54, 288–297
kinase. Mol. Cell Biol. 11, 5016–5031 Wu, C.W., Li, A.F., Chi, C.W., Lai, C.H., Huang, C.L., Lo, S.S.,
Papavassiliou, A.G., Treier, M. and Bohmann, D. (1995) Intramolecular Lui, W.Y. and Lin, W.C. (2002) Clinical significance of
signal transduction in c-Jun. EMBO J. 14, 2014–2019 AXL kinase family in gastric cancerAnticancer Res. 22,
Parekh, D.B., Ziegler, W. and Parker, P.J. (2000) Multiple pathways 1071–1078
control protein kinase C phosphorylation. EMBO J. 19, 496–503 Zenz, R., Scheuch, H., Martin, P., Frank, C., Eferl, R., Kenner, L.,
Pulverer, B.J., Kyriakis, J.M., Avruch, J., Nikolakaki, E. and Woodgett, Sibilia, M. and Wagner, E.F. (2003) c-Jun regulates eyelid closure
J.R. (1991) Phosphorylation of c-jun mediated by MAP kinases. and skin tumor development through EGFR signaling. Dev. Cell 4,
Nature 353, 670–674 879–889

Received 26 July 2010/11 October 2010; accepted 26 October 2010


Published as Immediate Publication 26 October 2010, doi:10.1042/BC20100094

www.biolcell.org | Volume 103 (1) | Pages 21–33 33


Biol. Cell (2011) 103, 21–33 (Printed in Great Britain) doi:10.1042/BC20100094

Supplementary online data


PMA up-regulates the transcription of Axl by AP-1 transcription
factor binding to TRE sequences via the MAPK cascade in
leukaemia cells
Giridhar Mudduluru∗ , Jörg H. Leupold∗ , Philipp Stroebel† and Heike Allgayer∗1
∗ Department of Experimental Surgery Mannheim/Molecular Oncology of Solid Tumors, DKFZ and University of Heidelberg, Mannheim
68167, Germany, and †Institute of Pathology, Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany

Figure S1 EMSA and Western blot for Sp family members


(a) EMSA analysis of 8 h PMA-stimulated or unstimulated nuclear extracts of K562 cells with Sp-binding motifs (Mudduluru
and Allgayer, 2008). (b) Sequential Western blot analysis for Sp1 and Sp3 was performed with PMA-stimulated K562 cells as
indicated. TPA, PMA.

1 Towhom correspondence should be addressed (email


heike.allgayer@umm.de).

www.biolcell.org | Volume 103 (1) | Pages 21–33


G. Mudduluru and others

Figure S2 AP-1 transcription factors bind to putative binding sites of the Axl promoter
EMSA was performed with or without PMA-stimulated nuclear extract of K562 cells and a γ-33 P-labelled oligonucleotide probe
corresponding to the GT-rich, AP-1 I, AP-1 II and AP-1 III sites of the Axl promoter. For competition, a 50-fold excess of
unlabelled wild-type or mutant oligonucleotides was used. The GT-rich region ( − 590/ − 610 bp) mutated for T with A, was
unable to abolish the binding of PMA-induced complexes [left-hand panel (GT-rich)], yet no specific binding complexes were
observed under unstimulated conditions. It shows that under stimulated conditions, PMA-induced transcription factors bind
to this GT-rich motif. Gel-shift analysis with AP-1 showed higher binding of specific complexes with PMA-stimulated samples
which were not observed in unstimulated conditions. Moreover, base conversion of GC into TT in this AP-1 I (middle pannel)
site failed to abolish the PMA-induced binding complexes when used as mutant competitor oligonucleotides. These results
showed that the complexes bound to AP-1 I motifs are highly specific. Higher binding of specific complexes bound to AP-1
II and III (consisting of TRE and CRE elements in close proximity in the promoter) were observed under PMA-stimulated
conditions (right-hand panel). Individual competition with oligonucleotides containing mutations in TRE elements, as well as
CRE elements, partially abolished the binding complexes; however, oligonucleotides containing mutations on both TRE
and CRE elements failed to do so. TPA, PMA.


C The Authors Journal compilation 
C 2011 Portland Press Limited
MAPK pathways in Axl gene expression

Figure S3 Supershift analysis of AP-1 family members


(a and b) Supershift analysis was performed as described in the Materials and methods section of the main text, with (bottom
panel) and without (top panel) 8 h PMA-stimulated nuclear extracts of K562, for AP-1 (c-Jun, Jun B, Jun D, c-Fos, Fos B, Fra
1 and Fra 2) using a GT-rich motif ( − 590/ − 610 bp) and AP-1 IV ( − 1013/ − 1019 bp) as radioactive probes. IgG served as the
negative control. Specific supershifted complexes are indicated. (c) Schematic presentation of an important region taken for
CHIP analysis. For, forward; Rev, reverse.

www.biolcell.org | Volume 103 (1) | Pages 21–33


G. Mudduluru and others

Figure S4 Western blot and supershift analysis for CREB


(a) Western blot analysis for phospho-CREB and total CREB was performed with nuclear extracts from 8 h PMA-stimulated
or unstimulated samples. DNA polymerase ε served as an internal control. (b) Supershift analysis of 8 h PMA-stimulated or
unstimulated nuclear extracts of K562 cells was performed by adding antibodies for phospho-CREB, CREB, and phospho-c-Jun
with AP-1 II and III motifs ( − 655/ − 640 bp) as the radioactive probe. IgG served as the negative control. Specific supershifted
complexes are indicated. TPA, PMA.


C The Authors Journal compilation 
C 2011 Portland Press Limited
MAPK pathways in Axl gene expression

Table S1 Primers used for EMSA and supershift analysis


The position is from the upstream translation start site. Lower-case letters represent the sequences used to mutate the respective
transcription-factor-binding motif. Letters in italics indicate the oligonucleotides used for EMSA analysis, apart from the respective
normal and mutated oligonucleotides.
Number Name Mutagenesis oligonucleotide sequences (5 →3 ) Position (bp)
1 Sp_a_For_N CAAAGGGGGAGCCAGGGGCGGAGAAAGGGTTGCCCAAG − 239 to − 202
2 Sp_a_For_M CAAAGGGGGAGCCAGGtttaGAGAAAGGGTTGCCCAAG
3 Sp_b_For_N GGCTCTGGCCCTGGTGGGCGGAGGCAAAGGGGGAGC − 263 to − 228
4 Sp_b_For_M GGCTCTGGCCCTGGTGGtttaAGGCAAAGGGGGAGC
5 Sp_c_For_N GGCTGGGGGTGGAGGCGGGGAGAGGGGCGTCACG − 524 to − 491
6 Sp_c_For_M GGCTGGGGGTGGAGtattGGAGAGGGGCGTCACG
7 Sp_d_For_N CCTGGCTGGGGCTGGGCTGGGGGTGGAGGCGGGGAGAGG − 538 to − 500
8 Sp_d_For_M CCTGGCTGGGGCTGGGtctttGGTGGAGGCGGGG AGAGG
9 Sp_e_For_N GGAGGCCTGGCTGGGGCTGGGCTGGGGGTGGAGGCG − 543 to − 508
10 Sp_e_For_M GGAGGCCTGGCTGGGtaTtaGCTGGGGGTGGAGGCG
11 Sp_cde_For_M CTGGCTGGGtattaGtctttGGTGGAGtattGGAGAGG − 537 to − 500
12 GT Rich_For_ N GTGTGTGTGTGTGTGTGTGTGTCCTTGTCCGAGGAGCCGA − 609 to − 569
13 GT Rich_For_M GAGAGAGAGAGAGAGAGAGAGACCTTGTCCGAGGAGCCGA
14 AP1 I_For_ N GTGTGTGTGCGTGCGTGCGTGCGTGCGTGTGTGT − 635 to − 601
15 AP1 I_For_ EM1 TTCTTTTTGCGTGCGTGCGTGCGTGCGTCTCTCT
16 AP1 I_For_ EM2 GTGTGTGTTTGTTTGTTTGTGCGTGCGTGTGTGT
17 AP1 I_For_ EM3 GTGTGTGTTTGTTTGTTTGTTTGTTTGTGTGTGT
18 AP1 I_For_ EM4 GTGTGTGTGCAAGCAAGCAAGCGTGCGTGTGTGT
19 AP1 I_For_ EM5 GTGTGTGTGCAAGCAAGCAAGCAAGCAAGTGTGT
20 AP1 I_For_ M1 CTAAGAGTGTGTGTGgaatCGTGCGTGCGTGCGTGTG − 641 to − 605
21 AP1 I_For_ M2 GTGTGTGgaatCGTGCGTGCaattGTGTGTGTGTGTGTGTGTGTG − 633 to − 589
22 AP1 I_For_ M3 CTAAGAGTGTGTGTGgaatCGattaaGCaattGTGTGTGTGTGTGTGTG − 641 to − 593
23 AP1 I II&III_For_ N GCTTGTCCTAGCCTGTGTGTGTCAGTGACGTCTAAGAGTGTG − 672 to − 631
24 AP1 I II_For_M CCTAGCCTGTGTGTGTCAGTttatTCTAAGAGTGTGTGTGCGTGCG − 666 to − 613
25 AP1 I III_For_M GCTTGTCCTAGCCTGTGcatacCAGTGACGTCTAAGAGTGTG − 672 to − 631
26 AP1 I II&III_For_M CCTAGCCTGTGcatacCAGTttatTCTAAGAGTGTGTGTGCGTGCG − 666 to − 613
27 AP1 IV_For_ N GTGCTGGGATTACAGGAGTGAGTTACTGTGCCCGGCCAAC − 1036 to − 1008
28 AP1 IV_For_ M GTGCTGGGATTACAGGAGTttcaTACTGTGCCCGGCCAAC

Table S2 Primers used for Axl promoter cloning


The position is from the upstream translation start site.
Number Name Oligonucleotide sequences (5 →3 ) Position (bp)
1 AxlB GAAGGTACCATGACAACCCAGGCAAAGTG − 2376
2 AxlP1 GAAGGTACCCAGTCCCACCAGAAGGAGAG − 1276
3 AxlP2 GAAGGTACCTGCCCGGCCAACAACTATTC − 1010
4 AxlP3 GAAGGTACCTGTCTGTGGGCCAGTAGC − 727
5 AxlP4 GAAGGTACCAATGAAGGGCCAAGGAGGC − 556
6 AxlP5 GAAGGTACCAGGGGAGTGGAGTTCTGG − 478
7 AxlP6 GAAGGTACCGGCAGGGGTGCTGAGAAG − 181
8 AxlD1 GAACTCGAGGCCTCCTTGGCCCTTCAT − 556
9 AxlD2 GAACTCGAGACCTCAGAGACAGGAGGACA − 770

www.biolcell.org | Volume 103 (1) | Pages 21–33


G. Mudduluru and others

Reference
Mudduluru, G. and Allgayer, H. (2008) The human receptor tyrosine
kinase Axl gene – promoter characterization and regulation of
constitutive expression by Sp1, Sp3 and CpG methylation. Biosci.
Rep. 28, 161–176

Received 26 July 2010/11 October 2010; accepted 26 October 2010


Published as Immediate Publication 26 October 2010, doi:10.1042/BC20100094


C The Authors Journal compilation 
C 2011 Portland Press Limited

You might also like